110 results on '"Shyue SK"'
Search Results
2. ADAPTIVE EVOLUTION OF COLOR-VISION GENES IN HIGHER PRIMATES
- Author
-
Shyue, Sk, Hewettemmett, D., Sperling, Hg, Hunt, Dm, Bowmaker, Jk, John Mollon, and Li, Wh
3. The effects of acetylated cordycepin derivatives on promoting vascular angiogenesis and attenuating myocardial ischemic injury.
- Author
-
Chang TC, Lin CF, Lu YJ, Liang SM, Wei JY, Chin CH, Shyue SK, Kuo CC, and Liou JY
- Abstract
Background: Enhanced angiogenesis following myocardial infarction (MI) is beneficial to preserve cardiac function. The present study aimed to investigate whether acetylated derivatives of cordycepin altered its original antitumor properties and exerted cardioprotective effects by promoting angiogenesis in vitro and in vivo ., Methods: Cordycepin and its derivatives with single (DA), double (DAA), and triple acetyl groups (DAAA) were assessed. The cell viability of leukemia U937 cells, malignant hepatoma Huh-7 cells, and human umbilical vascular endothelial cells (HUVECs) treated with cordycepin, DA, DAA, and DAAA were determined. The expression of β-catenin in U937 cells, as well as the expression of p65, p38 and other related signal regulators in HUVECs elicited by lipopolysaccharides (LPS) were also observed. Angiogenesis was determined by tube formation in HUVECs and Matrigel plug assay in mice. Cardiac function following administration of DAAA was evaluated in mice MI model simulated by coronary artery ligation., Results: The inhibitory effects of cordycepin and its acetylated derivatives on U937 cells, Huh-7 cells, HUVECs, and the expression of β-catenin in U937 cells were mitigated with increasing acetylation. Intriguingly, DAAA preserved the cell viability of HUVECs compared to other acetylated derivatives. Although DAAA had a significantly diminished antitumor effect compared to cordycepin, it promoted angiogenesis in mice and tube formation in HUVECs and attenuated LPS-induced phosphorylation of p65 and p38. Additionally, administration of DAAA improved cardiac function following coronary artery ligation in mice., Conclusion: DAAA could be considered a promising adjunctive therapy to prevent post-MI heart failure through promoting angiogenesis., Competing Interests: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (© 2024 The Authors.)
- Published
- 2024
- Full Text
- View/download PDF
4. Transient receptor potential vanilloid 1 inhibition reduces brain damage by suppressing neuronal apoptosis after intracerebral hemorrhage.
- Author
-
Chen CC, Ke CH, Wu CH, Lee HF, Chao Y, Tsai MC, Shyue SK, and Chen SF
- Subjects
- Animals, Mice, Male, Mice, Knockout, Capsaicin pharmacology, Capsaicin analogs & derivatives, Brain Injuries pathology, Brain Injuries metabolism, Brain Injuries drug therapy, Blood-Brain Barrier drug effects, Blood-Brain Barrier metabolism, Blood-Brain Barrier pathology, Brain pathology, Brain metabolism, Brain drug effects, TRPV Cation Channels metabolism, Cerebral Hemorrhage pathology, Cerebral Hemorrhage metabolism, Apoptosis drug effects, Neurons drug effects, Neurons metabolism, Neurons pathology, Mice, Inbred C57BL
- Abstract
Intracerebral hemorrhage (ICH) induces a complex sequence of apoptotic cascades and inflammatory responses, leading to neurological impairment. Transient receptor potential vanilloid 1 (TRPV1), a nonselective cation channel with high calcium permeability, has been implicated in neuronal apoptosis and inflammatory responses. This study used a mouse ICH model and neuronal cultures to examine whether TRPV1 activation exacerbates brain damage and neurological deficits by promoting neuronal apoptosis and neuroinflammation. ICH was induced by injecting collagenase in both wild-type (WT) C57BL/6 mice and TRPV1
-/- mice. Capsaicin (CAP; a TRPV1 agonist) or capsazepine (a TRPV1 antagonist) was administered by intracerebroventricular injection 30 min before ICH induction in WT mice. The effects of genetic deletion or pharmacological inhibition of TRPV1 using CAP or capsazepine on motor deficits, histological damage, apoptotic responses, blood-brain barrier (BBB) permeability, and neuroinflammatory reactions were explored. The antiapoptotic mechanisms and calcium influx induced by TRPV1 inactivation were investigated in cultured hemin-stimulated neurons. TRPV1 expression was upregulated in the hemorrhagic brain, and TRPV1 was expressed in neurons, microglia, and astrocytes after ICH. Genetic deletion of TRPV1 significantly attenuated motor deficits and brain atrophy for up to 28 days. Deletion of TRPV1 also reduced brain damage, neurodegeneration, microglial activation, cytokine expression, and cell apoptosis at 1 day post-ICH. Similarly, the administration of CAP ameliorated brain damage, neurodegeneration, brain edema, BBB permeability, and cytokine expression at 1 day post-ICH. In primary neuronal cultures, pharmacological inactivation of TRPV1 by CAP attenuated neuronal vulnerability to hemin-induced injury, suppressed apoptosis, and preserved mitochondrial integrity in vitro. Mechanistically, CAP reduced hemin-stimulated calcium influx and prevented the phosphorylation of CaMKII in cultured neurons, which was associated with reduced activation of P38 and c-Jun NH2 -terminal kinase mitogen-activated protein kinase signaling. Our results suggest that TRPV1 inhibition may be a potential therapy for ICH by suppressing mitochondria-related neuronal apoptosis., (© 2024 The Authors. Brain Pathology published by John Wiley & Sons Ltd on behalf of International Society of Neuropathology.)- Published
- 2024
- Full Text
- View/download PDF
5. Surf4 collaborates with derlin-2 and derlin-1 to mediate cyclooxygenase-2 translocation to the cytosol for degradation.
- Author
-
Chen SF, Wu CH, Lee YM, Tam K, Liou JY, and Shyue SK
- Abstract
Derlin family members participate in the retrotranslocation of endoplasmic reticulum (ER) lumen proteins to the cytosol for ER-associated degradation (ERAD); however, the proteins facilitating this retrotranslocation remain to be explored. Using CRISPR library screening, we have found that derlin-2 and surfeit locus protein 4 (Surf4) are candidates to facilitate degradation of cyclooxygenase-2 (COX-2, also known as PTGS2). Our results show that derlin-2 acts upstream of derlin-1 and that Surf4 acts downstream of derlin-2 and derlin-1 to facilitate COX-2 degradation. Knockdown of derlin-2 or Surf4 impedes the ubiquitylation of COX-2 and the interaction of COX-2 with caveolin-1 (Cav-1) and p97 (also known as VCP) in the cytosol. Additionally, COX-2 degradation is N-glycosylation dependent. Although derlin-2 facilitates degradation of N-glycosylated COX-2, the interaction between derlin-2 and COX-2 is independent of COX-2 N-glycosylation. Derlin-1, Surf4 and p97 preferentially interact with non-glycosylated COX-2, whereas Cav-1 preferentially interacts with N-glycosylated COX-2, regardless of the N-glycosylation pattern. Collectively, our results reveal that Surf4 collaborates with derlin-2 and derlin-1 to mediate COX-2 translocation from the ER lumen to the cytosol. The derlin-2-derlin-1-Surf4-Cav-1 machinery might represent a unique pathway to accelerate COX-2 degradation in ERAD., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2023. Published by The Company of Biologists Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
6. Peroxisome proliferator-activated receptor δ improves the features of atherosclerotic plaque vulnerability by regulating smooth muscle cell phenotypic switching.
- Author
-
Lien CF, Lin CS, Shyue SK, Hsieh PS, Chen SJ, Lin YT, Chien S, and Tsai MC
- Subjects
- Animals, Humans, Mice, Apolipoproteins E genetics, Apolipoproteins E metabolism, Inflammasomes metabolism, Matrix Metalloproteinase 2 metabolism, Mice, Inbred C57BL, Myocytes, Smooth Muscle metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Phenotype, Plaque, Atherosclerotic metabolism, PPAR delta genetics
- Abstract
Background and Purpose: Vascular smooth muscle cells (SMCs) undergo phenotypic switching during sustained inflammation, contributing to an unfavourable atherosclerotic plaque phenotype. PPARδ plays an important role in regulating SMC functions; however, its role in atherosclerotic plaque vulnerability remains unclear. Here, we explored the pathological roles of PPARδ in atherosclerotic plaque vulnerability in severe atherosclerosis and elucidated the underlying mechanisms., Experimental Approach: Plasma levels of PPARδ were measured in patients with acute coronary syndrome (ACS) and stable angina (SA). SMC contractile and synthetic phenotypic markers, endoplasmic reticulum (ER) stress, and features of atherosclerotic plaque vulnerability were analysed for the brachiocephalic artery of apolipoprotein E-knockout (ApoE
-/- ) mice, fed a high-cholesterol diet (HCD) and treated with or without the PPARδ agonist GW501516. In vitro, the role of PPARδ was elucidated using human aortic SMCs (HASMCs)., Key Results: Patients with ACS had significantly lower plasma PPARδ levels than those with SA. GW501516 reduced atherosclerotic plaque vulnerability, a synthetic SMC phenotype, ER stress markers, and NLRP3 inflammasome expression in HCD-fed ApoE-/- mice. ER stress suppressed PPARδ expression in HASMCs. PPARδ activation inhibited ER stress-induced synthetic phenotype development, ER stress-NLRP3 inflammasome axis activation and matrix metalloproteinase 2 (MMP2) expression in HASMCs. PPARδ inhibited NFκB signalling and alleviated ER stress-induced SMC phenotypic switching., Conclusions and Implications: Low plasma PPARδ levels may be associated with atherosclerotic plaque vulnerability. Our findings provide new insights into the mechanisms underlying the protective effect of PPARδ on SMC phenotypic switching and improvement the features of atherosclerotic plaque vulnerability., (© 2023 British Pharmacological Society.)- Published
- 2023
- Full Text
- View/download PDF
7. The Role of SCL Isoforms in Embryonic Hematopoiesis.
- Author
-
Chuang CK, Chen SF, Su YH, Chen WH, Lin WM, Wang IC, and Shyue SK
- Subjects
- Mice, Animals, Embryonic Development genetics, Embryo, Mammalian metabolism, Endothelium, Endothelial Cells, Hematopoiesis genetics
- Abstract
Three waves of hematopoiesis occur in the mouse embryo. The primitive hematopoiesis appears as blood islands in the extra embryonic yolk sac at E7.5. The extra embryonic pro-definitive hematopoiesis launches in late E8 and the embryonic definitive one turns on at E10.5 indicated by the emergence of hemogenic endothelial cells on the inner wall of the extra embryonic arteries and the embryonic aorta. To study the roles of SCL protein isoforms in murine hematopoiesis, the SCL-large (SCL-L) isoform was selectively destroyed with the remaining SCL-small (SCL-S) isoform intact. It was demonstrated that SCL-S was specifically expressed in the hemogenic endothelial cells (HECs) and SCL-L was only detected in the dispersed cells after budding from HECs. The SCL
Δ/Δ homozygous mutant embryos only survived to E10.5 with normal extra embryonic vessels and red blood cells. In wild-type mouse embryos, a layer of neatly aligned CD34+ and CD43+ cells appeared on the endothelial wall of the aorta of the E10.5 fetus. However, the cells at the same site expressed CD31 rather than CD34 and/or CD43 in the E10.5 SCLΔ/Δ embryo, indicating that only the endothelial lineage was developed. These results reveal that the SCL-S is sufficient to sustain the primitive hematopoiesis and SCL-L is necessary to launch the definitive hematopoiesis.- Published
- 2023
- Full Text
- View/download PDF
8. Fibroblasts Drive Metabolic Reprogramming in Pacemaker Cardiomyocytes.
- Author
-
Chou PC, Liu CM, Weng CH, Yang KC, Cheng ML, Lin YC, Yang RB, Shyu BC, Shyue SK, Liu JD, Chen SP, Hsiao M, and Hu YF
- Subjects
- Animals, Cellular Reprogramming, Coculture Techniques, Fibroblasts metabolism, Mice, Rats, Sinoatrial Node metabolism, Induced Pluripotent Stem Cells metabolism, Myocytes, Cardiac metabolism
- Abstract
Background: The sino atrial node (SAN) is characterized by the microenvironment of pacemaker cardiomyocytes (PCs) encased with fibroblasts. An altered microenvironment leads to rhythm failure. Operable cell or tissue models are either generally lacking or difficult to handle. The biological process behind the milieu of SANs to evoke pacemaker rhythm is unknown. We explored how fibroblasts interact with PCs and regulate metabolic reprogramming and rhythmic activity in the SAN., Methods: Tbx18 (T-box transcription factor 18)-induced PCs and fibroblasts were used for cocultures and engineered tissues, which were used as the in vitro models to explore how fibroblasts regulate the functional integrity of SANs. RNA-sequencing, metabolomics, and cellular and molecular techniques were applied to characterize the molecular signals underlying metabolic reprogramming and identify its critical regulators. These pathways were further validated in vivo in rodents and induced human pluripotent stem cell-derived cardiomyocytes., Results: We observed that rhythmicity in Tbx18-induced PCs was regulated by aerobic glycolysis. Fibroblasts critically activated metabolic reprogramming and aerobic glycolysis within PCs, and, therefore, regulated pacemaker activity in PCs. The metabolic reprogramming was attributed to the exclusive induction of Aldoc (aldolase c) within PCs after fibroblast-PC integration. Fibroblasts activated the integrin-dependent mitogen-activated protein kinase-E2F1 signal through cell-cell contact and turned on Aldoc expression in PCs. Interruption of fibroblast-PC interaction or Aldoc knockdown nullified electrical activity. Engineered Tbx18-PC tissue sheets were generated to recapitulate the microenvironment within SANs. Aldoc-driven rhythmic machinery could be replicated within tissue sheets. Similar machinery was faithfully validated in de novo PCs of adult mice and rats, and in human PCs derived from induced pluripotent stem cells., Conclusions: Fibroblasts drive Aldoc-mediated metabolic reprogramming and rhythmic regulation in SANs. This work details the cellular machinery behind the complex milieu of vertebrate SANs and opens a new direction for future therapy.
- Published
- 2022
- Full Text
- View/download PDF
9. Atypical antipsychotic drugs deregulate the cholesterol metabolism of macrophage-foam cells by activating NOX-ROS-PPARγ-CD36 signaling pathway.
- Author
-
Chen CH, Leu SJ, Hsu CP, Pan CC, Shyue SK, and Lee TS
- Subjects
- Animals, Atherosclerosis pathology, Foam Cells metabolism, Mice, Mice, Knockout, Olanzapine pharmacology, PPAR gamma metabolism, Antipsychotic Agents pharmacology, CD36 Antigens metabolism, Cholesterol metabolism, Foam Cells drug effects, NADPH Oxidases metabolism, Reactive Oxygen Species metabolism
- Abstract
Background: Clinical reports indicate that schizophrenia patients taking atypical antipsychotic drugs suffer from metabolism diseases including atherosclerosis. However, the mechanisms underlying the detrimental effect of atypical antipsychotic drugs on atherosclerosis remain to be explored., Methods: In this study, we used apolipoprotein E-deficient (apoe
-/- ) hyperlipidemic mice and apoe-/- cd36-/- mice to investigate the underlying mechanism of atypical antipsychotic drugs on atherosclerosis and macrophage-foam cells., Results: In vivo studies showed that genetic deletion of cd36 gene ablated the pro-atherogenic effect of olanzapine in apoe-/- mice. Moreover, in vitro studies revealed that genetic deletion or siRNA-mediated knockdown of cd36 or pharmacological inhibition of CD36 prevented atypical antipsychotic drugs-induced oxLDL accumulation in macrophages. Additionally, olanzapine and clozapine activated NADPH oxidase (NOX) to generate reactive oxygen species (ROS) which upregulated the activity of peroxisome proliferator-activated receptor γ (PPARγ) and subsequently elevated CD36 expression. Inhibition of NOX activity, ROS production or PPARγ activity suppressed CD36 expression and abolished the detrimental effects of olanzapine and clozapine on oxLDL accumulation in macrophages., Conclusion: Collectively, our results suggest that atypical antipsychotic drugs exacerbate atherosclerosis and macrophage-foam cell formation by activating the NOX-ROS-PPARγ-CD36 pathway., Competing Interests: Declaration of competing interest The authors declare no conflict of interest., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
10. The phosphatase activity of soluble epoxide hydrolase regulates ATP-binding cassette transporter-A1-dependent cholesterol efflux.
- Author
-
Lien CC, Chen CH, Lee YM, Guo BC, Cheng LC, Pan CC, Shyue SK, and Lee TS
- Subjects
- ATP Binding Cassette Transporter 1 metabolism, Animals, Apolipoproteins E deficiency, Atherosclerosis genetics, Atherosclerosis metabolism, Epoxide Hydrolases antagonists & inhibitors, Foam Cells metabolism, Lipid Metabolism, Lipoproteins, LDL metabolism, Macrophages metabolism, Mice, Mice, Inbred C57BL, Phosphorylation, Protein Binding, ATP Binding Cassette Transporter 1 genetics, Atherosclerosis enzymology, Cholesterol metabolism, Epoxide Hydrolases genetics, Epoxide Hydrolases metabolism, Foam Cells enzymology, Macrophages enzymology, Phosphoprotein Phosphatases metabolism
- Abstract
The contribution of soluble epoxide hydrolase (sEH) to atherosclerosis has been well defined. However, less is understood about the role of sEH and its underlying mechanism in the cholesterol metabolism of macrophages. The expression of sEH protein was increased in atherosclerotic aortas of apolipoprotein E-deficient mice, primarily in macrophage foam cells. Oxidized low-density lipoprotein (oxLDL) increased sEH expression in macrophages. Genetic deletion of sEH (sEH
-/- ) in macrophages markedly exacerbated oxLDL-induced lipid accumulation and decreased the expression of ATP-binding cassette transporters-A1 (ABCA1) and apolipoprotein AI-dependent cholesterol efflux following oxLDL treatment. The down-regulation of ABCA1 in sEH-/- macrophages was due to an increase in the turnover rate of ABCA1 protein but not in mRNA transcription. Inhibition of phosphatase activity, but not hydrolase activity, of sEH decreased ABCA1 expression and cholesterol efflux following oxLDL challenge, which resulted in increased cholesterol accumulation. Additionally, oxLDL increased the phosphatase activity, promoted the sEH-ABCA1 complex formation and decreased the phosphorylated level of ABCA1 at threonine residues. Overexpression of phosphatase domain of sEH abrogated the oxLDL-induced ABCA1 phosphorylation and further increased ABCA1 expression and cholesterol efflux, leading to the attenuation of oxLDL-induced cholesterol accumulation. Our findings suggest that the phosphatase domain of sEH plays a crucial role in the cholesterol metabolism of macrophages., (© 2019 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine.)- Published
- 2019
- Full Text
- View/download PDF
11. Activation of TrkB/Akt signaling by a TrkB receptor agonist improves long-term histological and functional outcomes in experimental intracerebral hemorrhage.
- Author
-
Wu CH, Chen CC, Hung TH, Chuang YC, Chao M, Shyue SK, and Chen SF
- Subjects
- Animals, Cerebral Hemorrhage chemically induced, Collagenases toxicity, Injections, Intraperitoneal, Male, Mice, Mice, Inbred C57BL, Protein-Tyrosine Kinases, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, Cerebral Hemorrhage drug therapy, Flavones pharmacology, Membrane Glycoproteins agonists, Neuroprotective Agents pharmacology, Signal Transduction drug effects
- Abstract
Background: Intracerebral hemorrhage (ICH) induces a complex sequence of apoptotic cascades that contribute to secondary neuronal damage. Tropomyosin-related kinase receptor B (TrkB) signaling plays a crucial role in promoting neuronal survival following brain damage., Methods: The present study investigated the protective effects and underlying mechanisms of TrkB activation by the specific TrkB agonist, 7,8-dihydroxyflavone (7,8-DHF), in a model of collagenase-induced ICH and in neuronal cultures. Mice subjected to collagenase-induced ICH were intraperitoneally injected with either 7,8-DHF or vehicle 10 min after ICH and, subsequently, daily for 3 days. Behavioral studies, brain edema measurement, and histological analysis were conducted. Levels of TrkB signaling-related molecules and apoptosis-related proteins were analyzed by western blots., Results: Treatment with 20 mg/kg 7,8-DHF significantly improved functional recovery and reduced brain damage up to 28 days post-ICH. Reduction in neuronal death, apoptosis, and brain edema were also observed in response to 7,8-DHF treatment at 3 days post-ICH. These changes were accompanied by a significant increase in the phosphorylation of TrkB and Akt (Ser473/Thr308) at 1 and 3 days, but had no effect on Erk 44/42 phosphorylation. 7,8-DHF also enhanced the phosphorylation of Ask-1 Ser967 and FOXO-1, downstream targets of Akt at 1 and 3 days. Moreover, 7,8-DHF increased brain-derived neurotrophic factor levels at 1 day. In primary cultured neurons stimulated with hemin, 7,8-DHF promoted survival and reduced apoptosis. Furthermore, delaying the administration of 7,8-DHF to 3 h post-ICH reduced brain tissue damage and neuronal death., Conclusions: Our findings demonstrate that the activation of TrkB signaling by 7,8-DHF protects against ICH via the Akt, but not the Erk, pathway. These data provide new insights into the role of TrkB signaling deficit in the pathophysiology of ICH and highlight TrkB/Akt as possible therapeutic targets in this disease.
- Published
- 2019
- Full Text
- View/download PDF
12. ZNF479 downregulates metallothionein-1 expression by regulating ASH2L and DNMT1 in hepatocellular carcinoma.
- Author
-
Wu YJ, Ko BS, Liang SM, Lu YJ, Jan YJ, Jiang SS, Shyue SK, Chen L, and Liou JY
- Subjects
- 14-3-3 Proteins genetics, 14-3-3 Proteins metabolism, Animals, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular pathology, Cell Proliferation drug effects, Cell Proliferation genetics, DNA (Cytosine-5-)-Methyltransferase 1 genetics, DNA-Binding Proteins genetics, Elafin antagonists & inhibitors, Elafin genetics, Elafin metabolism, Hep G2 Cells, Histones metabolism, Humans, Liver Neoplasms genetics, Liver Neoplasms pathology, MAP Kinase Signaling System drug effects, MAP Kinase Signaling System genetics, Metallothionein genetics, Methylation, Mice, Mice, Inbred BALB C, Mice, Nude, Nuclear Proteins genetics, TOR Serine-Threonine Kinases antagonists & inhibitors, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases metabolism, Transcription Factors genetics, Transplantation, Heterologous, Carcinoma, Hepatocellular metabolism, DNA (Cytosine-5-)-Methyltransferase 1 metabolism, DNA-Binding Proteins metabolism, Liver Neoplasms metabolism, Metallothionein metabolism, Nuclear Proteins metabolism, Transcription Factors metabolism
- Abstract
Decreased expression of metallothionein-1 (MT-1) is associated with a poor prognosis in hepatocellular carcinoma (HCC). Here, we found that MT-1 expression was suppressed by 14-3-3ε, and MT-1 overexpression abolished 14-3-3ε-induced cell proliferation and tumor growth. We identified that 14-3-3ε induced expression of ZNF479, a novel potential transcriptional regulator by gene expression profiling and ZNF479 contributed to 14-3-3ε-suppressed MT-1 expression. ZNF479 induced the expression of DNMT1, UHRF1, and mixed-lineage leukemia (MLL) complex proteins (ASH2L and Menin), and increased tri-methylated histone H3 (H3K4me3) levels, but suppressed H3K4 (H3K4me2) di-methylation. ZNF479-suppressed MT-1 expression was restored by silencing of ASH2L and DNMT1. Furthermore, ZNF479 expression was higher in HCC tissues than that in the non-cancerous tissues. Expression analyses revealed a positive correlation between the expression of ZNF479 and DNMT1, UHRF1, ASH2L, and Menin, and an inverse correlation with that of ZNF479, ASH2L, Menin, and MT-1 isoforms. Moreover, correlations between the expression of ZNF479 and its downstream factors were more pronounced in HCC patients with hepatitis B. Here, we found that ZNF479 regulates MT-1 expression by modulating ASH2L in HCC. Approaches that target ZNF479/MLL complex/MT-1 or related epigenetic regulatory factors are potential therapeutic strategies for HCC.
- Published
- 2019
- Full Text
- View/download PDF
13. Cordycepin Suppresses Endothelial Cell Proliferation, Migration, Angiogenesis, and Tumor Growth by Regulating Focal Adhesion Kinase and p53.
- Author
-
Lin YT, Liang SM, Wu YJ, Wu YJ, Lu YJ, Jan YJ, Ko BS, Chuang YJ, Shyue SK, Kuo CC, and Liou JY
- Abstract
Focal adhesion kinase (FAK) plays an important role in vascular development, including the regulation of endothelial cell (EC) adhesion, migration, proliferation, and survival. 3'-deoxyadenosine (cordycepin) is known to suppress FAK expression, cell migration, and the epithelial⁻mesenchymal transition in hepatocellular carcinoma (HCC). However, whether cordycepin affects FAK expression and cellular functions in ECs and the specific molecular mechanism remain unclear. In this study, we found that cordycepin suppressed FAK expression and the phosphorylation of FAK (p-FAK) at Tyr397 in ECs. Cordycepin inhibited the proliferation, wound healing, transwell migration, and tube formation of ECs. Confocal microscopy revealed that cordycepin significantly reduced FAK expression and decreased focal adhesion number of ECs. The suppressed expression of FAK was accompanied by induced p53 and p21 expression in ECs. Finally, we demonstrated that cordycepin suppressed angiogenesis in an in vivo angiogenesis assay and reduced HCC tumor growth in a xenograft nude mice model. Our study indicated that cordycepin could attenuate cell proliferation and migration and may result in the impairment of the angiogenesis process and tumor growth via downregulation of FAK and induction of p53 and p21 in ECs. Therefore, cordycepin may be used as a potential adjuvant for cancer therapy.
- Published
- 2019
- Full Text
- View/download PDF
14. Deletion of caveolin-1 attenuates LPS/GalN-induced acute liver injury in mice.
- Author
-
Tsai TH, Tam K, Chen SF, Liou JY, Tsai YC, Lee YM, Huang TY, and Shyue SK
- Subjects
- Animals, Apoptosis genetics, Chemical and Drug Induced Liver Injury pathology, Hepatocytes drug effects, Hepatocytes metabolism, Humans, Inflammation chemically induced, Inflammation pathology, Intercellular Adhesion Molecule-1 genetics, Kupffer Cells drug effects, Kupffer Cells metabolism, Lipopolysaccharide Receptors genetics, Lipopolysaccharides toxicity, Liver injuries, Liver metabolism, Liver pathology, Mice, NF-kappa B genetics, Neutrophil Infiltration genetics, Nitric Oxide Synthase Type II genetics, Signal Transduction genetics, Toll-Like Receptor 4 genetics, Transcription Factor RelA genetics, Caveolin 1 genetics, Chemical and Drug Induced Liver Injury genetics, Inflammation genetics, Liver drug effects
- Abstract
Acute hepatic injury caused by inflammatory liver disease is associated with high mortality. This study examined the role of caveolin-1 (Cav-1) in lipopolysaccharide (LPS) and D-galactosamine (GalN)-induced fulminant hepatic injury in wild type and Cav-1-null (Cav-1
-/- ) mice. Hepatic Cav-1 expression was induced post-LPS/GalN treatment in wild-type mice. LPS/GalN-treated Cav-1-/- mice showed reduced lethality and markedly attenuated liver damage, neutrophil infiltration and hepatocyte apoptosis as compared to wild-type mice. Cav-1 deletion significantly reduced LPS/GalN-induced caspase-3, caspase-8 and caspase-9 activation and pro-inflammatory cytokine and chemokine expression. Additionally, Cav-1-/- mice showed suppressed expression of Toll-like receptor 4 (TLR4) and CD14 in Kupffer cells and reduced expression of vascular cell adhesion molecule 1 and intercellular adhesion molecule 1 in liver cells. Cav-1 deletion impeded LPS/GalN-induced inducible nitric oxide synthase expression and nitric oxide production and hindered nuclear factor-κB (NF-κB) activation. Taken together, Cav-1 regulated the expression of mediators that govern LPS-induced inflammatory signalling in mouse liver. Thus, deletion of Cav-1 suppressed the inflammatory response mediated by the LPS-CD14-TLR4-NF-κb pathway and alleviated acute liver injury in mice., (© 2018 Institute of Biomedical Sciences, Academia Sinica. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine.)- Published
- 2018
- Full Text
- View/download PDF
15. Correction: Regulation of Aldo-keto-reductase family 1 B10 by 14-3-3ε and their prognostic impact of hepatocellular carcinoma.
- Author
-
Liu TA, Jan YJ, Ko BS, Wu YJ, Lu YJ, Liang SM, Liu CC, Chen SC, Wang J, Shyue SK, and Liou JY
- Abstract
[This corrects the article DOI: 10.18632/oncotarget.5734.].
- Published
- 2018
- Full Text
- View/download PDF
16. Atypical Antipsychotic Drug Olanzapine Deregulates Hepatic Lipid Metabolism and Aortic Inflammation and Aggravates Atherosclerosis.
- Author
-
Chen CH, Shyue SK, Hsu CP, and Lee TS
- Subjects
- Adipose Tissue, White pathology, Animals, Aorta drug effects, Aorta pathology, Apolipoproteins E deficiency, Apolipoproteins E genetics, Atherosclerosis metabolism, Atherosclerosis veterinary, Blood Pressure drug effects, Cholesterol analysis, Cholesterol blood, Fatty Acids analysis, Hyperlipidemias metabolism, Hyperlipidemias pathology, Hyperlipidemias veterinary, Inflammation, Liver drug effects, Liver pathology, Male, Mice, Mice, Knockout, Olanzapine, Triglycerides blood, Antipsychotic Agents pharmacology, Aorta metabolism, Atherosclerosis pathology, Benzodiazepines pharmacology, Lipid Metabolism drug effects, Liver metabolism
- Abstract
Background/aims: Olanzapine, an atypical antipsychotic drug, has therapeutic effects for schizophrenia. However, clinical reports indicate that patients taking atypical antipsychotic drugs are at high risk of metabolic syndrome with unclear mechanisms. We investigated the effect of olanzapine on atherosclerosis and the mechanisms in apolipoprotein E-null (apoE-/-) mice., Methods: ApoE-/- mice were used as in vivo models. Western blot analysis was used to evaluate protein expression. Conventional assay kits were applied to assess the levels of cholesterol, triglycerides, free cholesterol, cholesteryl ester, fatty acids, glycerol, and cytokines., Results: Daily treatment with olanzapine (3 mg/kg body weight) for four weeks increased mean arterial blood pressure and the whitening of brown adipose tissue in mice. In addition, olanzapine impaired aortic cholesterol homeostasis and exacerbated hyperlipidemia and aortic inflammation, which accelerated atherosclerosis in mice. Moreover, lipid accumulation in liver, particularly total cholesterol, free cholesterol, fatty acids, and glycerol, was increased with olanzapine treatment in apoE-/- mice by upregulating the expression of de novo lipid synthesis-related proteins and downregulating that of cholesterol clearance- or very low-density lipoprotein secretion-related proteins., Conclusion: Olanzapine may exacerbate atherosclerosis by deregulating hepatic lipid metabolism and worsening hyperlipidemia and aortic inflammation., (© 2018 The Author(s). Published by S. Karger AG, Basel.)
- Published
- 2018
- Full Text
- View/download PDF
17. Genetic deletion or pharmacological inhibition of soluble epoxide hydrolase reduces brain damage and attenuates neuroinflammation after intracerebral hemorrhage.
- Author
-
Wu CH, Shyue SK, Hung TH, Wen S, Lin CC, Chang CF, and Chen SF
- Subjects
- Animals, Brain Injuries etiology, Brain Injuries pathology, Cerebral Hemorrhage complications, Cerebral Hemorrhage enzymology, Inflammation etiology, Inflammation pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Brain Injuries enzymology, Cerebral Hemorrhage pathology, Epoxide Hydrolases metabolism, Inflammation enzymology
- Abstract
Background: Inflammatory responses significantly contribute to neuronal damage and poor functional outcomes following intracerebral hemorrhage (ICH). Soluble epoxide hydrolase (sEH) is known to induce neuroinflammatory responses via degradation of anti-inflammatory epoxyeicosatrienoic acids (EET), and sEH is upregulated in response to brain injury. The present study investigated the involvement of sEH in ICH-induced neuroinflammation, brain damage, and functional deficits using a mouse ICH model and microglial cultures., Methods: ICH was induced by injecting collagenase in both wild-type (WT) C57BL/6 mice and sEH knockout (KO) mice. WT mice were injected intracerebroventricularly with 12-(3-adamantan-1-yl-ureido)-dodecanoic acid (AUDA), a selective sEH inhibitor, 30 min before ICH. Expression of sEH in the hemorrhagic hemisphere was examined by immunofluorescence and Western blot analysis. The effects of genetic deletion or pharmacological inhibition of sEH by AUDA on neuroinflammatory responses, EET degradation, blood-brain barrier (BBB) permeability, histological damage, and functional deficits were evaluated. The anti-inflammatory mechanism of sEH inactivation was investigated in thrombin- or hemin-stimulated cultured microglia., Results: ICH induced an increase in sEH protein levels in the hemorrhagic hemisphere from 3 h to 4 days. sEH was expressed in microglia/macrophages, astrocytes, neurons, and endothelial cells in the perihematomal region. Genetic deletion of sEH significantly attenuated microglia/macrophage activation and expression of inflammatory mediators and reduced EET degradation at 1 and 4 days post-ICH. Deletion of sEH also reduced BBB permeability, matrix metalloproteinase (MMP)-9 activity, neutrophil infiltration, and neuronal damage at 1 and 4 days. Likewise, administration of AUDA attenuated proinflammatory microglia/macrophage activation and EET degradation at 1 day post-ICH. These findings were associated with a reduction in functional deficits and brain damage for up to 28 days. AUDA also ameliorated neuronal death, BBB disruption, MMP-9 activity, and neutrophil infiltration at 1 day. However, neither gene deletion nor pharmacological inhibition of sEH altered the hemorrhage volume following ICH. In primary microglial cultures, genetic deletion or pharmacological inhibition of sEH by AUDA reduced thrombin- and hemin-induced microglial activation. Furthermore, AUDA reduced thrombin- and hemin-induced P38 MAPK and NF-κB activation in BV2 microglia cultures. Ultimately, AUDA attenuated N2A neuronal death that was induced by BV2 microglial conditioned media., Conclusions: Our results suggest that inhibition of sEH may provide a potential therapy for ICH by suppressing microglia/macrophage-mediated neuroinflammation.
- Published
- 2017
- Full Text
- View/download PDF
18. Deletion or inhibition of soluble epoxide hydrolase protects against brain damage and reduces microglia-mediated neuroinflammation in traumatic brain injury.
- Author
-
Hung TH, Shyue SK, Wu CH, Chen CC, Lin CC, Chang CF, and Chen SF
- Abstract
Traumatic brain injury (TBI) induces a series of inflammatory processes that contribute to neuronal damage. The present study investigated the involvement of soluble epoxide hydrolase (sEH) in neuroinflammation and brain damage in mouse TBI and in microglial cultures. The effects of genetic deletion of sEH and treatment with an sEH inhibitor, 12-(3-adamantan-1-yl-ureido)-dodecanoic acid (AUDA), on brain damage and inflammatory responses were evaluated in mice subjected to controlled cortical impact. The anti-inflammatory mechanism of sEH inhibition/deletion was investigated in vitro . TBI-induced an increase in sEH protein level in the injured cortex from 1 h to 4 days and sEH was expressed in microglia. Genetic deletion of sEH significantly attenuated functional deficits and brain damage up to 28 days post-TBI. Deletion of sEH also reduced neuronal death, apoptosis, brain edema, and BBB permeability at 1 and 4 day(s). These changes were associated with markedly reduced microglial/macrophage activation, neutrophil infiltration, matrix metalloproteinase-9 activity, inflammatory mediator expression at 1 and 4 day(s), and epoxyeicosatrienoic acid (EET) degradation at 1 and 4 day(s). Administration of AUDA attenuated brain edema, apoptosis, inflammatory mediator upregulation and EET degradation at 4 days. In primary microglial cultures, AUDA attenuated both LPS- or IFN-γ-stimulated nitric oxide (NO) production and reduced LPS- or IFN-γ-induced p38 MAPK and NF-κB signaling. Deletion of sEH also reduced IFN-γ-induced NO production. Moreover, AUDA attenuated N2A neuronal death induced by BV2 microglial-conditioned media. Our results suggest that inhibition of sEH may be a potential therapy for TBI by modulating the cytotoxic functions of microglia., Competing Interests: CONFLICTS OF INTEREST The authors declare that they have no conflicts of interest.
- Published
- 2017
- Full Text
- View/download PDF
19. Corrigendum: Role of phosphatase activity of soluble epoxide hydrolase in regulating simvastatin-activated endothelial nitric oxide synthase.
- Author
-
Hou HH, Liao YJ, Hsiao SH, Shyue SK, and Lee TS
- Abstract
This corrects the article DOI: 10.1038/srep13524.
- Published
- 2017
- Full Text
- View/download PDF
20. Improving the regenerative potential of olfactory ensheathing cells by overexpressing prostacyclin synthetase and its application in spinal cord repair.
- Author
-
Tsai MJ, Huang CT, Huang YS, Weng CF, Shyue SK, Huang MC, Liou DY, Lin YR, Cheng CH, Kuo HS, Lin Y, Lee MJ, Huang WH, Huang WC, and Cheng H
- Subjects
- Animals, Cells, Cultured, Cytochrome P-450 Enzyme System metabolism, Intramolecular Oxidoreductases metabolism, Rats, Rats, Sprague-Dawley, Recovery of Function, Cytochrome P-450 Enzyme System genetics, Gene Expression, Intramolecular Oxidoreductases genetics, Neuroglia physiology, Olfactory Nerve physiology, Spinal Cord Regeneration
- Abstract
Background: Olfactory ensheathing cells (OEC), specialized glia that ensheathe bundles of olfactory nerves, have been reported as a favorable substrate for axonal regeneration. Grafting OEC to injured spinal cord appears to facilitate axonal regeneration although the functional recovery is limited. In an attempt to improve the growth-promoting properties of OEC, we transduced prostacyclin synthase (PGIS) to OEC via adenoviral (Ad) gene transfer and examined the effect of OEC with enhanced prostacyclin synthesis in co-culture and in vivo. Prostacyclin is a vasodilator, platelet anti-aggregatory and cytoprotective agent., Results: Cultured OEC expressed high level of cyclooxygneases, but not PGIS. Infection of AdPGIS to OEC could selectively augument prostacyclin synthesis. When cocultured with either OEC or AdPGIS-OEC, neuronal cells were resistant to OGD-induced damage. The resulted OEC were further transplanted to the transected cavity of thoracic spinal cord injured (SCI) rats. By 6 weeks post-surgery, significant functional recovery in hind limbs occurred in OEC or AdPGIS-OEC transplanted SCI rats compared with nontreated SCI rats. At 10-12 weeks postgraft, AdPGIS-OEC transplanted SCI rats showed significantly better motor restoration than OEC transplanted SCI rats. Futhermore, regenerating fiber tracts in the distal spinal cord stump were found in 40-60% of AdPGIS-OEC transplanted SCI rats., Conclusions: Enhanced synthesis of prostacyclin in grafted OEC improved fiber tract regeneration and functional restoration in spinal cord injured rats. These results suggest an important potential of prostacyclin in stimulating OEC therapeutic properties that are relevant for neural transplant therapies.
- Published
- 2017
- Full Text
- View/download PDF
21. Cordycepin disrupts leukemia association with mesenchymal stromal cells and eliminates leukemia stem cell activity.
- Author
-
Liang SM, Lu YJ, Ko BS, Jan YJ, Shyue SK, Yet SF, and Liou JY
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Cell Adhesion drug effects, Deoxyadenosines administration & dosage, Disease Models, Animal, Humans, K562 Cells, Leukemia pathology, Mice, Survival Analysis, Treatment Outcome, U937 Cells, Antineoplastic Agents pharmacology, Deoxyadenosines pharmacology, Leukemia drug therapy, Mesenchymal Stem Cells drug effects, Mesenchymal Stem Cells physiology
- Abstract
Maintaining stemness of leukemic stem cells (LSCs) and reciprocal interactions between leukemia and stromal cells support leukemic progression and resistance to chemotherapy. Targeting the niche-based microenvironment is thus a new approach for leukemia therapy. Cordycepin is an analogue of adenosine and has been suggested to possess anti-leukemia properties. However, whether cordycepin influences association of leukemia and mesenchymal stromal cells has never been investigated. Here we show that cordycepin reduces CD34
+ CD38- cells in U937 and K562 cells and induces Dkk1 expression via autocrine and paracrine regulation in leukemia and mesenchymal stromal/stem cells (MSCs). Cordycepin suppresses cell attachment of leukemia with MSCs and downregulates N-cadherin in leukemia and VCAM-1 in MSCs. Moreover, incubation with leukemic conditioned media (CM) significantly induces IL-8 and IL-6 expression in MSCs, which is abrogated by cordycepin. Suppression of leukemic CM-induced VCAM-1 and IL-8 by cordycepin in MSCs is mediated by impairing NFκB signaling. Finally, cordycepin combined with an adenosine deaminase inhibitor prolongs survival in a leukemic mouse model. Our results indicate that cordycepin is a potential anti-leukemia therapeutic adjuvant via eliminating LSCs and disrupting leukemia-stromal association.- Published
- 2017
- Full Text
- View/download PDF
22. Pyrogallol abates VSMC migration via modulation of Caveolin-1, matrix metalloproteinase and intima hyperplasia in carotid ligation mouse.
- Author
-
Ma YD, Thiyagarajan V, Tsai MJ, Lue SI, Chia YC, Shyue SK, and Weng CF
- Subjects
- Animals, Carotid Arteries pathology, Caveolin 1 genetics, Cell Culture Techniques, Cell Survival drug effects, Cells, Cultured, Disease Models, Animal, Male, Matrix Metalloproteinases genetics, Meliaceae chemistry, Mice, Inbred C57BL, Mice, Knockout, Myocytes, Smooth Muscle metabolism, Myocytes, Smooth Muscle pathology, Neointima etiology, Neointima metabolism, Neointima pathology, Pyrogallol isolation & purification, Pyrogallol pharmacology, Tunica Intima drug effects, Tunica Intima pathology, Carotid Arteries drug effects, Caveolin 1 metabolism, Cell Movement drug effects, Matrix Metalloproteinases metabolism, Myocytes, Smooth Muscle drug effects, Neointima prevention & control, Pyrogallol therapeutic use
- Abstract
Migration of vascular smooth muscle cells (VSMCs) contributes to intimal hyperplasia and other vascular diseases. Caveolin-1 (Cav-1) has been recognized as a proliferative inhibitor of VSMCs and is likely to be an important regulator of VSMC migration. The underlying mechanism of pyrogallol on the VSMC migration is not fully understood. This study attempted to dissect the role of Cav-1 and matrix metalloproteinase (MMP) in VSMC migration and to investigate the effect of pyrogallol on VSMC mobility during carotid artery ligation mice. The mRNA expression of MMP-3 and MMP-13 was down-regulated in cultured VSMC prepared from Cav-1-deficient (Cav-1 KO) mice whereas MMP-14 expression was up-regulated. Pyrogallol effectively inhibited the migration of Cav-1 KO VSMC by promoting the expression of tissue inhibitors of metalloproteinase (TIMP)-2. Pyrogallol also inhibited the migration of Cav-1 wild type (WT) VSMC, however, by increasing TIMP-1 expression and repressing MMP-2 activity. In a parallel in vivo study, intra-peritoneal (ip) of pyrogallol to carotid artery ligated mice significantly suppressed intima formation in mice carotid artery. Furthermore, the proMMP-9 activity in pyrogallol-treated mice serum significantly increased from Day 0 to Day 2 and decreased from Day 2 to Day 7 in a time-dependent manner. In addition, WT mice treated with pyrogallol had significantly reduced neointima formation, whereas no differences were observed in Cav-1 knock out (KO) mice. These results suggest that pyrogallol not only inhibited VSMC migration but also effectively diminishes the severity of neointima hyperplasia, implying that pyrogallol possesses potential anti-atherogenic effects for the treatment of vascular diseases., (Copyright © 2016 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
23. Transient Receptor Potential Ankyrin 1 Channel Involved in Atherosclerosis and Macrophage-Foam Cell Formation.
- Author
-
Zhao JF, Shyue SK, Kou YR, Lu TM, and Lee TS
- Subjects
- Animals, Apolipoproteins E genetics, Apolipoproteins E metabolism, Atherosclerosis genetics, Blotting, Western, Cholesterol metabolism, Foam Cells drug effects, Immunohistochemistry, Isothiocyanates therapeutic use, Lipoproteins, LDL pharmacology, Macrophages drug effects, Mice, Mice, Inbred C57BL, Mice, Knockout, NF-kappa B pharmacology, TRPA1 Cation Channel, Transient Receptor Potential Channels antagonists & inhibitors, Atherosclerosis metabolism, Foam Cells cytology, Foam Cells metabolism, Macrophages cytology, Macrophages metabolism, Transient Receptor Potential Channels metabolism
- Abstract
Transient receptor potential ankyrin 1 channel (TRPA1) plays an important role in the pathogenesis of inflammatory diseases, yet its role and the underlying mechanism in atherosclerosis remain unclear. We aimed to investigate the role of TRPA1 in atherosclerosis and foam-cell formation in vivo in mice and in vitro in mouse macrophages. Histopathology was examined by hematoxylin and eosin staining, levels of cytokines and lipid profile were evaluated by assay kits, and protein expression was determined by western blot analysis. TRPA1 expression was increased in macrophage foam cells in atherosclerotic aortas of apolipoprotein E-deficient (apoE(-/-)) mice. Atherosclerotic lesions, hyperlipidemia and systemic inflammation were worsened with chronic administration of the TRPA1 channel antagonist HC030031 or genetic ablation of TRPA1 (TRPA1(-/-)) in apoE(-/-) mice. Treatment with allyl isothiocyanate (AITC, a TRPA1 agonist) retarded the progression of atherosclerosis in apoE(-/-) mice but not apoE(-/-)TRPA1(-/-) mice. Mouse macrophages showed oxidized low-density lipoprotein (oxLDL) activated TRPA1 channels. OxLDL-induced lipid accumulation of macrophages was exacerbated by HC030031 or loss of function of TRPA1. Inhibition of TRPA1 activity did not alter oxLDL internalization but impaired cholesterol efflux by downregulating the ATP-binding cassette transporters. Furthermore, tumor necrosis factor-α-induced inflammatory response was attenuated in AITC-activated macrophages. TRPA1 may be a pivotal regulator in the pathogenesis of atherosclerosis and cholesterol metabolism of macrophage foam cells.
- Published
- 2016
- Full Text
- View/download PDF
24. Maternal exposure to di-(2-ethylhexyl) phthalate exposure deregulates blood pressure, adiposity, cholesterol metabolism and social interaction in mouse offspring.
- Author
-
Lee KI, Chiang CW, Lin HC, Zhao JF, Li CT, Shyue SK, and Lee TS
- Subjects
- Animals, Biomarkers blood, Brain drug effects, Brain pathology, Brain physiopathology, Female, Hypertension physiopathology, Liver drug effects, Liver metabolism, Maze Learning drug effects, Mice, Inbred C57BL, Motor Activity drug effects, Pregnancy, Adiposity drug effects, Behavior, Animal drug effects, Blood Pressure drug effects, Cholesterol blood, Diethylhexyl Phthalate toxicity, Hypertension chemically induced, Maternal Exposure, Prenatal Exposure Delayed Effects, Social Behavior
- Abstract
Long-term exposure to di-(2-ethylhexyl) phthalate (DEHP) is highly associated with carcinogenicity, fetotoxicity, psychological disorders and metabolic diseases, but the detrimental effects and mechanisms are not fully understood. We investigated the effect of exposing mouse mothers to DEHP, and the underlying mechanism, on blood pressure, obesity and cholesterol metabolism as well as psychological and learning behaviors in offspring. Tail-cuff plethysmography was used for blood pressure measurement; Western blot used was for phosphorylation and expression of protein; hematoxylin and eosin staining, Nissl staining and Golgi staining were used for histological examination. The serum levels of cholesterol, triglycerides and glucose were measured by blood biochemical analysis. Hepatic cholesterol and triglyceride levels were assessed by colorimetric assay kits. Offspring behaviors were evaluated by open-field activity, elevated plus maze, social preference test and Morris water maze. Maternal DEHP exposure deregulated the phosphorylation of endothelial nitric oxide synthase and upregulated angiotensin type 1 receptor in offspring, which led to increased blood pressure. It led to obesity in offspring by increasing the size of adipocytes in white adipose tissue and number of adipocytes in brown adipose tissue. It increased the serum level of cholesterol in offspring by decreasing the hepatic capacity for cholesterol clearance. The impaired social interaction ability induced by maternal DEHP exposure might be due to abnormal neuronal development. Collectively, our findings provide new evidence that maternal exposure to DEHP has a lasting effect on the physiological functions of the vascular system, adipose tissue and nerve system in offspring.
- Published
- 2016
- Full Text
- View/download PDF
25. Role of transient receptor potential ankyrin 1 channels in Alzheimer's disease.
- Author
-
Lee KI, Lee HT, Lin HC, Tsay HJ, Tsai FC, Shyue SK, and Lee TS
- Subjects
- Animals, Behavior, Animal, Blotting, Western, Brain metabolism, Disease Models, Animal, Immunohistochemistry, Mice, Mice, Inbred C57BL, Mice, Transgenic, Signal Transduction physiology, TRPA1 Cation Channel, Alzheimer Disease metabolism, Astrocytes metabolism, Transient Receptor Potential Channels metabolism
- Abstract
Background: Transient receptor potential ankyrin 1 (TRPA1) channel plays an important role in pain and inflammation. However, little is known about the significance of the TRPA1 channel in the pathophysiology of Alzheimer's disease (AD)., Methods: Wild-type (WT), TRPA1(-/-), amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic (APP/PS1 Tg) mice, the mouse model of AD, and APP/PS1 Tg/TRPA1(-/-) mice were used to examine the role of TRPA1 in pathogenesis of AD. Western blot was used for protein expression; immunohistochemistry was used for histological examination. The mouse behaviors were evaluated by locomotion, nesting building, Y-maze and Morris water maze tests; levels of interleukin (IL)-1β, IL-4, IL-6 and IL-10 and the activities of protein phosphatase 2B (PP2B), NF-κB and nuclear factor of activated T cells (NFAT) were measured by conventional assay kits; Fluo-8 NW calcium (Ca(2+)) assay kit was used for the measurement of intracellular Ca(2+) level in primary astrocytes and HEK293 cells., Results: The protein expression of TRPA1 channels was higher in brains, mainly astrocytes of the hippocampus, from APP/PS1 Tg mice than WT mice. Ablation of TRPA1-channel function in APP/PS1 Tg mice alleviated behavioral dysfunction, Aβ plaque deposition and pro-inflammatory cytokine production but increased astrogliosis in brain lesions. TRPA1 channels were activated and Ca(2+) influx was elicited in both astrocytes and TRPA1-transfected HEK293 cells treated with fibrilized Aβ1-42; these were abrogated by pharmacological inhibition of TRPA1 channel activity, disruption of TRPA1 channel function or removal of extracellular Ca(2+). Inhibition of TRPA1 channel activity exacerbated Aβ1-42-induced astrogliosis but inhibited Aβ1-42-increased PP2B activation, the production of pro-inflammatory cytokines and activities of transcriptional factors NF-κB and NFAT in astrocytes and in APP/PS1 Tg mice. Pharmacological inhibition of PP2B activity diminished the fibrilized Aβ1-42-induced production of pro-inflammatory cytokines, activation of NF-κB and NFAT and astrogliosis in astrocytes., Conclusions: TRPA1 - Ca(2+) - PP2B signaling may play a crucial role in regulating astrocyte-derived inflammation and pathogenesis of AD.
- Published
- 2016
- Full Text
- View/download PDF
26. Asymmetric Dimethylarginine Limits the Efficacy of Simvastatin Activating Endothelial Nitric Oxide Synthase.
- Author
-
Hsu CP, Zhao JF, Lin SJ, Shyue SK, Guo BC, Lu TM, and Lee TS
- Subjects
- Aged, Animals, Arginine blood, Cardiovascular Diseases prevention & control, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Endothelium, Vascular metabolism, Enzyme Activation drug effects, Female, Humans, Male, Mice, Nitric Oxide biosynthesis, Nitric Oxide Synthase Type III metabolism, Reactive Oxygen Species metabolism, Signal Transduction drug effects, Arginine analogs & derivatives, Nitric Oxide Synthase Type III drug effects, Simvastatin therapeutic use
- Abstract
Background: Asymmetric dimethylarginine (ADMA), an endogenous inhibitor of endothelial nitric oxide synthase (eNOS), is considered a risk factor for the pathogenesis of cardiovascular diseases. Simvastatin, a lipid-lowering drug with other pleiotropic effects, has been widely used for treatment of cardiovascular diseases. However, little is known about the effect and underlying molecular mechanisms of ADMA on the effectiveness of simvastatin in the vascular system., Methods and Results: We conducted a prospective cohort study to enroll 648 consecutive patients with coronary artery disease for a follow-up period of 8 years. In patients with plasma ADMA level ≥0.49 μmol/L (a cut-off value from receiver operating characteristic curve), statin treatment had no significant effect on cardiovascular events. We also conducted randomized, controlled studies using in vitro and in vivo models. In endothelial cells, treatment with ADMA (≥0.5 μmol/L) impaired simvastatin-induced nitric oxide (NO) production, endothelial NO synthase (eNOS) phosphorylation, and angiogenesis. In parallel, ADMA markedly increased the activity of NADPH oxidase (NOX) and production of reactive oxygen species (ROS). The detrimental effects of ADMA on simvastatin-induced NO production and angiogenesis were abolished by the antioxidant, N-acetylcysteine, NOX inhibitor, or apocynin or overexpression of dimethylarginine dimethylaminohydrolase 2 (DDAH-2). Moreover, in vivo, ADMA administration reduced Matrigel plug angiogenesis in wild-type mice and decreased simvastatin-induced eNOS phosphorylation in aortas of apolipoprotein E-deficient mice, but not endothelial DDAH-2-overexpressed aortas., Conclusions: We conclude that ADMA may trigger NOX-ROS signaling, which leads to restricting the simvastatin-conferred protection of eNOS activation, NO production, and angiogenesis as well as the clinical outcome of cardiovascular events., (© 2016 The Authors. Published on behalf of the American Heart Association, Inc., by Wiley Blackwell.)
- Published
- 2016
- Full Text
- View/download PDF
27. The GroEL protein of Porphyromonas gingivalis regulates atherogenic phenomena in endothelial cells mediated by upregulating toll-like receptor 4 expression.
- Author
-
Huang CY, Shih CM, Tsao NW, Lin YW, Shih CC, Chiang KH, Shyue SK, Chang YJ, Hsieh CK, and Lin FY
- Abstract
Porphyromonas gingivalis (P. gingivalis) is a bacterial species that causes periodontitis. GroEL from P. gingivalis may possess biological activity and may be involved in the destruction of periodontal tissues. However, it is unclear whether P. gingivalis GroEL enhances the appearance of atherogenic phenomena in endothelial cells and vessels. Here, we constructed recombinant GroEL from P. gingivalis to investigate its effects in human coronary artery endothelial cells (HCAECs) in vitro and on aortas of high-cholesterol (HC)-fed B57BL/6 and B57BL/6-Tlr4(lps-del) mice in vivo. The results showed that GroEL impaired tube-formation capacity under non-cytotoxic conditions in HCAECs. GroEL increased THP-1 cell/HCAEC adhesion by increasing the expression of intracellular adhesion molecule (ICAM)-1 and vascular adhesion molecule (VCAM)-1 in endothelial cells. Additionally, GroEL increased DiI-oxidized low density lipoprotein (oxLDL) uptake, which may be mediated by elevated lectin-like oxLDL receptor (LOX)-1 but not scavenger receptor expressed by endothelial cells (SREC) and scavenger receptor class B1 (SR-B1) expression. Furthermore, GroEL interacts with toll-like receptor 4 (TLR4) and plays a causal role in atherogenesis in HCAECs. Human antigen R (HuR), an RNA-binding protein with a high affinity for the 3' untranslated region (3'UTR) of TLR4 mRNA, contributes to the up-regulation of TLR4 induced by GroEL in HCAECs. In a GroEL animal administration study, GroEL elevated ICAM-1, VCAM-1, LOX-1 and TLR4 expression in the aortas of HC diet-fed wild C57BL/6 but not C57BL/6-Tlr4(lps-del) mice. Taken together, our findings suggest that P. gingivalis GroEL may contribute to cardiovascular disorders by affecting TLR4 expression.
- Published
- 2016
28. Excess Nitric Oxide Activates TRPV1-Ca(2+)-Calpain Signaling and Promotes PEST-dependent Degradation of Liver X Receptor α.
- Author
-
Zhao JF, Shyue SK, and Lee TS
- Subjects
- Animals, Biological Transport drug effects, Blotting, Western, Cell Line, Foam Cells drug effects, Foam Cells metabolism, Humans, Immunoprecipitation, Liver X Receptors, Macrophages drug effects, Macrophages metabolism, Mice, Nitric Oxide pharmacology, Polyamines pharmacology, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, S-Nitroso-N-Acetylpenicillamine pharmacology, Signal Transduction drug effects, ATP Binding Cassette Transporter 1 metabolism, Calcium metabolism, Calpain metabolism, Nitric Oxide metabolism, Orphan Nuclear Receptors metabolism
- Abstract
Excess nitric oxide (NO) deregulates cholesterol metabolism in macrophage foam cells, yet the underlying molecular mechanism is incompletely understood. To investigate the mechanism, we found that in macrophages, treatment with NO donors S-nitroso-N-acetyl-D,L-penicillamine (SNAP) or diethylenetriamine/nitric oxide induced LXRα degradation and reduced the expression of the downstream target of LXRα, ATP-binding cassette transporter A1 (ABCA1), and cholesterol efflux. In addition, SNAP induced calcium (Ca(2+)) influx into cells, increased calpain activity and promoted the formation of calpain-LXRα complex. Pharmacological inhibition of calpain activity reversed the SNAP-induced degradation of LXRα, down-regulation of ABCA1 and impairment of cholesterol efflux in macrophages. SNAP increased the formation of calpain-LXRα complex in a Pro-Glu-Ser-Thr (PEST) motif-dependent manner. Truncation of the PEST motif in LXRα abolished the calpain-dependent proteolysis. Removal of extracellular Ca(2+) by EGTA or pharmacological inhibition of TRPV1 channel activity diminished SNAP-induced increase in intracellular Ca(2+), calpain activation, LXRα degradation, ABCA1 down-regulation and impaired cholesterol efflux. In conclusion, excess NO may activate calpain via TRPV1-Ca(2+) signaling and promote the recognition of calpain in the PEST motif of LXRα, thereby leading to degradation of LXRα and, ultimately, downregulated ABCA1 expression and impaired ABCA1-dependent cholesterol efflux in macrophages.
- Published
- 2016
- Full Text
- View/download PDF
29. Di-(2-ethylhexyl) phthalate accelerates atherosclerosis in apolipoprotein E-deficient mice.
- Author
-
Zhao JF, Hsiao SH, Hsu MH, Pao KC, Kou YR, Shyue SK, and Lee TS
- Subjects
- Adipose Tissue, White drug effects, Adipose Tissue, White metabolism, Adiposity drug effects, Animals, Apolipoproteins E genetics, Atherosclerosis blood, Atherosclerosis genetics, Atherosclerosis pathology, Cell Line, Cholesterol blood, Disease Models, Animal, Disease Progression, Endothelial Cells drug effects, Endothelial Cells metabolism, Genetic Predisposition to Disease, Humans, Inflammation Mediators blood, Lipoproteins, LDL blood, Liver drug effects, Liver metabolism, Macrophages drug effects, Macrophages metabolism, Male, Mice, Knockout, Phenotype, Risk Assessment, Time Factors, Apolipoproteins E deficiency, Atherosclerosis chemically induced, Diethylhexyl Phthalate toxicity, Plasticizers toxicity
- Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is associated with atherosclerosis-related cardiovascular disease complications, but we lack direct evidence of its unfavorable effect on atherogenesis. In this study, we aimed to clarify in vivo and in vitro the contribution of DEHP to the development of atherosclerosis and its underlying mechanisms. Apolipoprotein E-deficient (apoE(-/-)) mice chronically treated with DEHP for 4 weeks showed exacerbated hyperlipidemia, systemic inflammation, and atherosclerosis. In addition, DEHP promoted low-density lipoprotein (LDL) oxidation, which led to inflammation in endothelial cells as evidenced by increased protein expression of pro-inflammatory mediators. Furthermore, chronic DEHP treatment increased hepatic cholesterol accumulation by downregulating the protein expression of key regulators in cholesterol clearance including LDL receptor, cholesterol 7α-hydrolase, ATP-binding cassette transporter G5 and G8, and liver X receptor α. Moreover, the adiposity and inflammation of white adipose tissues were promoted in DEHP-treated apoE(-/-) mice. In conclusion, DEHP may disturb cholesterol homeostasis and deregulate the inflammatory response, thus leading to accelerated atherosclerosis.
- Published
- 2016
- Full Text
- View/download PDF
30. Rho-associated kinase inhibitors promote the cardiac differentiation of embryonic and induced pluripotent stem cells.
- Author
-
Cheng YT, Yeih DF, Liang SM, Chien CY, Yu YL, Ko BS, Jan YJ, Kuo CC, Sung LY, Shyue SK, Chen MF, Yet SF, Wu KK, and Liou JY
- Subjects
- 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine analogs & derivatives, 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine pharmacology, Amides pharmacology, Animals, Basic Helix-Loop-Helix Transcription Factors metabolism, Biomarkers metabolism, Cell Differentiation drug effects, Echocardiography, Embryonic Stem Cells cytology, Embryonic Stem Cells enzymology, Gene Expression, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells drug effects, Hematopoietic Stem Cells metabolism, Induced Pluripotent Stem Cells cytology, Male, Mice, Myocardial Infarction therapy, Myocytes, Cardiac cytology, Platelet Membrane Glycoprotein IIb metabolism, Pluripotent Stem Cells cytology, Pluripotent Stem Cells enzymology, Pyridines pharmacology, Real-Time Polymerase Chain Reaction methods, Stem Cell Transplantation methods, rho-Associated Kinases metabolism, Embryonic Stem Cells drug effects, Induced Pluripotent Stem Cells drug effects, Myocytes, Cardiac drug effects, Pluripotent Stem Cells drug effects, Protein Kinase Inhibitors pharmacology, rho-Associated Kinases antagonists & inhibitors
- Abstract
Background: Rho-associated kinase (ROCK) plays an important role in maintaining embryonic stem (ES) cell pluripotency. To determine whether ROCK is involved in ES cell differentiation into cardiac and hematopoietic lineages, we evaluated the effect of ROCK inhibitors, Y-27632 and fasudil on murine ES and induced pluripotent stem (iPS) cell differentiation., Methods: Gene expression levels were determined by real-time PCR, Western blot analysis and immunofluorescent confocal microscopy. Cell transplantation of induced differentiated cells were assessed in vivo in a mouse model (three groups, n=8/group) of acute myocardial infarction (MI). The cell engraftment was examined by immunohistochemical staining and the outcome was analyzed by echocardiography., Results: Cells were cultured in hematopoietic differentiation medium in the presence or absence of ROCK inhibitor and colony formation as well as markers of ES, hematopoietic stem cells (HSC) and cells of cardiac lineages were analyzed. ROCK inhibition resulted in a drastic change in colony morphology accompanied by loss of hematopoietic markers (GATA-1, CD41 and β-Major) and expressed markers of cardiac lineages (GATA-4, Isl-1, Tbx-5, Tbx-20, MLC-2a, MLC-2v, α-MHC, cTnI and cTnT) in murine ES and iPS cells. Fasudil-induced cardiac progenitor (Mesp-1 expressing) cells were infused into a murine MI model. They engrafted into the peri-infarct and infarct regions and preserved left ventricular function., Conclusions: These findings provide new insights into the signaling required for ES cell differentiation into hematopoietic as well as cardiac lineages and suggest that ROCK inhibitors are useful in directing iPS cell differentiation into cardiac progenitor cells for cell therapy of cardiovascular diseases., (Copyright © 2015 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
31. Regulation of aldo-keto-reductase family 1 B10 by 14-3-3ε and their prognostic impact of hepatocellular carcinoma.
- Author
-
Liu TA, Jan YJ, Ko BS, Wu YJ, Lu YJ, Liang SM, Liu CC, Chen SC, Wang J, Shyue SK, and Liou JY
- Subjects
- 14-3-3 Proteins genetics, Carcinoma, Hepatocellular enzymology, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular pathology, Cell Line, Tumor, Cell Proliferation physiology, Female, Humans, Liver Neoplasms enzymology, Liver Neoplasms genetics, Liver Neoplasms pathology, Male, Prognosis, Signal Transduction, 14-3-3 Proteins metabolism, Carcinoma, Hepatocellular metabolism, Liver Neoplasms metabolism
- Abstract
14-3-3ε is overexpressed in hepatocellular carcinoma (HCC) and its expression significantly associates with a poor prognostic outcome. To uncover how 14-3-3ε contributes to the tumor progression of HCC, we investigated the potential downstream targets regulated by 14-3-3ε. We found that 14-3-3ε increases expression and nuclear translocation of β-catenin and that 14-3-3ε-induced cell proliferation is attenuated by β-catenin silencing in HCC cells. Moreover, 14-3-3ε induces aldo-keto reductase family 1 member B10 (AKR1B10) expression through the activation of β-catenin signaling. Knockdown of AKR1B10 by siRNAs abolished 14-3-3ε-induced in vitro cell proliferation, anchorage-independent growth as well as in vivo tumor growth. Furthermore, AKR1B10 silencing increased retinoic acid (RA) levels in the serum of tumor-bearing mice and RA treatment attenuated 14-3-3ε-induced HCC cell proliferation. We further examined 14-3-3ε and AKR1B10 expression and clinicopathological characteristics of HCC tumors. Although the expression of AKR1B10 was significantly correlated with 14-3-3ε, an increase of AKR1B10 expression in 14-3-3ε positive patients paradoxically had better overall survival and disease-free survival rates as well as lower metastatic incidence than those without an AKR1B10 increase. Finally, we found a loss of AKR1B10 expression in cells exhibiting a high capacity of invasiveness. Silencing of AKR1B10 resulted in inducing snail and vimentin expression in HCC cells. These results indicate that AKR1B10 may play a dual role during HCC tumor progression. Our results also indicate that 14-3-3ε regulates AKR1B10 expression by activating β-catenin signaling. A combination of 14-3-3ε with AKR1B10 is a potential therapeutic target and novel prognostic biomarker of HCC.
- Published
- 2015
- Full Text
- View/download PDF
32. TRC8 downregulation contributes to the development of non-alcoholic steatohepatitis by exacerbating hepatic endoplasmic reticulum stress.
- Author
-
Chang PC, Tsai HW, Chiang MT, Huang PL, Shyue SK, and Chau LY
- Abstract
Endoplasmic reticulum (ER) stress is implicated in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). TRC8 is an ER-resident E3 ligase with roles in modulating lipid and protein biosynthesis. In this study we showed that TRC8 expression was downregulated in steatotic livers of patients and mice fed with a high fat diet (HFD) or a methionine and choline deficient (MCD) diet. To investigate the impact of TRC8 downregulation on steatosis and the progression to non-alcoholic steatohepatitis (NASH), we placed TRC8 knockout (KO) mice and wild type (WT) controls on a HFD or MCD diet and the severities of steatosis and NASH developed were compared. We found that TRC8 deficiency did not significantly affect diet-induced steatosis. Nevertheless, MCD diet-induced NASH as characterized by hepatocyte death, inflammation and fibrosis were exacerbated in TRC8-KO mice. The hepatic ER stress response, as evidenced by increased eIF2α phosphorylation and expression of ATF4 and CHOP, and the level of activated caspase 3, an apoptosis indicator, were augmented by TRC8 deficiency. The hepatic ER stress and NASH induced in mice could be ameliorated by adenovirus-mediated hepatic TRC8 overexpression. Mechanistically, we found that TRC8 deficiency augmented lipotoxic-stress-induced unfolded protein response in hepatocytes by attenuating the arrest of protein translation and the misfolded protein degradation. These findings disclose a crucial role of TRC8 in the maintenance of ER protein homeostasis and its downregulation in steatotic liver contributes to the progression of NAFLD., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
33. Growth arrest DNA damage-inducible gene 45 gamma expression as a prognostic and predictive biomarker in hepatocellular carcinoma.
- Author
-
Ou DL, Shyue SK, Lin LI, Feng ZR, Liou JY, Fan HH, Lee BS, Hsu C, and Cheng AL
- Subjects
- Aged, Animals, Antineoplastic Agents pharmacology, Apoptosis drug effects, Blotting, Western, CCAAT-Enhancer-Binding Proteins genetics, CCAAT-Enhancer-Binding Proteins metabolism, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular mortality, Cell Cycle Proteins analysis, Cell Cycle Proteins genetics, Cell Line, Tumor, Chromatin Immunoprecipitation, Female, Gene Expression Regulation, Neoplastic genetics, Humans, Kaplan-Meier Estimate, Liver Neoplasms genetics, Liver Neoplasms mortality, Male, Mice, Mice, Inbred BALB C, Middle Aged, Niacinamide analogs & derivatives, Niacinamide pharmacology, Nuclear Proteins analysis, Nuclear Proteins genetics, Phenylurea Compounds pharmacology, Prognosis, Proportional Hazards Models, RNA, Small Interfering, Reverse Transcriptase Polymerase Chain Reaction, Sorafenib, Transfection, Xenograft Model Antitumor Assays, Biomarkers, Tumor analysis, Carcinoma, Hepatocellular pathology, Cell Cycle Proteins biosynthesis, Drug Resistance, Neoplasm genetics, Liver Neoplasms pathology, Nuclear Proteins biosynthesis
- Abstract
Growth arrest DNA damage-inducible gene 45 (GADD45) family proteins play a crucial role in regulating cellular stress responses and apoptosis. The present study explored the prognostic and predictive role of GADD45γ in hepatocellular carcinoma (HCC) treatment. GADD45γ expression in HCC cells was examined using quantitative reverse transcription-PCR (qRT-PCR) and Western blotting. The control of GADD45γ transcription was examined using a luciferase reporter assay and chromatin immunoprecipitation. The in vivo induction of GADD45γ was performed using adenoviral transfer. The expression of GADD45γ in HCC tumor tissues from patients who had undergone curative resection was measured using qRT-PCR. Sorafenib induced expression of GADD45γ mRNA and protein, independent of its RAF kinase inhibitor activity. GADD45γ induction was more prominent in sorafenib-sensitive HCC cells (Huh-7 and HepG2, IC50 6-7 μM) than in sorafenib-resistant HCC cells (Hep3B, Huh-7R, and HepG2R, IC50 12-15 μM). Overexpression of GADD45γ reversed sorafenib resistance in vitro and in vivo, whereas GADD45γ expression knockdown by using siRNA partially abrogated the proapoptotic effects of sorafenib on sorafenib-sensitive cells. Overexpression of survivin in HCC cells abolished the antitumor enhancement between GADD45γ overexpression and sorafenib treatment, suggesting that survivin is a crucial mediator of antitumor effects of GADD45γ. GADD45γ expression decreased in tumors from patients with HCC who had undergone curative surgery, and low GADD45γ expression was an independent prognostic factor for poor survival, in addition to old age and vascular invasion. The preceding data indicate that GADD45γ suppression is a poor prognostic factor in patients with HCC and may help predict sorafenib efficacy in HCC.
- Published
- 2015
- Full Text
- View/download PDF
34. Role of phosphatase activity of soluble epoxide hydrolase in regulating simvastatin-activated endothelial nitric oxide synthase.
- Author
-
Hou HH, Liao YJ, Hsiao SH, Shyue SK, and Lee TS
- Subjects
- Dose-Response Relationship, Drug, Endothelial Cells drug effects, Enzyme Activation drug effects, Solubility, Endothelial Cells enzymology, Epoxide Hydrolases metabolism, Nitric Oxide Synthase Type III metabolism, Phosphoric Monoester Hydrolases metabolism, Simvastatin administration & dosage
- Abstract
Soluble epoxide hydrolase (sEH) has C-terminal epoxide hydrolase and N-terminal lipid phosphatase activity. Its hydrolase activity is associated with endothelial nitric oxide synthase (eNOS) dysfunction. However, little is known about the role of sEH phosphatase in regulating eNOS activity. Simvastatin, a clinical lipid-lowering drug, also has a pleiotropic effect on eNOS activation. However, whether sEH phosphatase is involved in simvastatin-activated eNOS activity remains elusive. We investigated the role of sEH phosphatase activity in simvastatin-mediated activation of eNOS in endothelial cells (ECs). Simvastain increased the phosphatase activity of sEH, which was diminished by pharmacological inhibitors of sEH phosphatase. In addition, pharmacological inhibition of sEH phosphatase or overexpressing the inactive phosphatase domain of sEH enhanced simvastatin-induced NO bioavailability, tube formation and phosphorylation of eNOS, Akt, and AMP-activated protein kinase (AMPK). In contrast, overexpressing the phosphatase domain of sEH limited the simvastatin-increased NO biosynthesis and eNOS phosphorylation at Ser1179. Simvastatin evoked epidermal growth factor receptor-c-Src-increased Tyr phosphorylation of sEH and formation of an sEH-Akt-AMPK-eNOS complex, which was abolished by the c-Src kinase inhibitor PP1 or c-Src dominant-negative mutant K298M. These findings suggest that sEH phosphatase activity negatively regulates simvastatin-activated eNOS by impeding the Akt-AMPK-eNOS signaling cascade.
- Published
- 2015
- Full Text
- View/download PDF
35. The GroEL protein of Porphyromonas gingivalis accelerates tumor growth by enhancing endothelial progenitor cell function and neovascularization.
- Author
-
Lin FY, Huang CY, Lu HY, Shih CM, Tsao NW, Shyue SK, Lin CY, Chang YJ, Tsai CS, Lin YW, and Lin SJ
- Subjects
- Allantois blood supply, Animals, Cell Line, Tumor, Chick Embryo, E-Selectin metabolism, Endothelial Progenitor Cells cytology, Humans, Male, Mice, Mice, Inbred BALB C, Microscopy, Confocal, Neovascularization, Physiologic, Nitric Oxide Synthase Type III metabolism, Phosphorylation, Porphyromonas gingivalis genetics, Recombinant Proteins metabolism, Virulence Factors metabolism, p38 Mitogen-Activated Protein Kinases metabolism, Bacterial Proteins metabolism, Chaperonin 60 metabolism, Colonic Neoplasms microbiology, Endothelial Progenitor Cells metabolism, Porphyromonas gingivalis pathogenicity
- Abstract
Porphyromonas gingivalis is a bacterial species that causes destruction of periodontal tissues. Additionally, previous evidence indicates that GroEL from P. gingivalis may possess biological activities involved in systemic inflammation, especially inflammation involved in the progression of periodontal diseases. The literature has established a relationship between periodontal disease and cancer. However, it is unclear whether P. gingivalis GroEL enhances tumor growth. Here, we investigated the effects of P. gingivalis GroEL on neovasculogenesis in C26 carcinoma cell-carrying BALB/c mice and chick eggs in vivo as well as its effect on human endothelial progenitor cells (EPC) in vitro. We found that GroEL treatment accelerated tumor growth (tumor volume and weight) and increased the mortality rate in C26 cell-carrying BALB/c mice. GroEL promoted neovasculogenesis in chicken embryonic allantois and increased the circulating EPC level in BALB/c mice. Furthermore, GroEL effectively stimulated EPC migration and tube formation and increased E-selectin expression, which is mediated by eNOS production and p38 mitogen-activated protein kinase activation. Additionally, GroEL may enhance resistance against paclitaxel-induced cell cytotoxicity and senescence in EPC. In conclusion, P. gingivalis GroEL may act as a potent virulence factor, contributing to the neovasculogenesis of tumor cells and resulting in accelerated tumor growth., (© 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
36. Linear correlation between average fluorescence intensity of green fluorescent protein and the multiplicity of infection of recombinant adenovirus.
- Author
-
Tsai YC, Tsai TH, Chang CP, Chen SF, Lee YM, and Shyue SK
- Subjects
- Cell Line, Tumor, Cytomegalovirus physiology, Flow Cytometry, Genetic Vectors genetics, Genetic Vectors metabolism, Humans, Phosphoglycerate Kinase metabolism, Adenoviridae physiology, Adenoviridae Infections virology, Fluorescence, Green Fluorescent Proteins metabolism, Promoter Regions, Genetic
- Abstract
Background: Adenoviral vector is an efficient tool for gene transfer. Protein expression is regulated by a number of factors, but the regulation by gene copy number remains to be investigated further., Results: Assessed by flow cytometry, we demonstrated a significant linear correlation between average fluorescence intensity of green fluorescent protein (GFP) and a wide range of multiplicity of infection (MOI), spanning from 0.01 to 200. Average GFP intensity was calculated by mean fluorescence intensity (MFI) × percentage of infection (POI) (MFI × POI) and the correlation was observed in cells transduced with GFP-expressing adenoviral vector driven either by a cytomegalovirus (CMV) promoter for 3 to 6 h or by a human phosphoglycerate kinase (PGK) promoter for 18 to 24 h. Factors impacting this linear correlation include MOI of viral vector, strength of promoter driving GFP expression, cell type transduced and incubation time after gene transfer. We also found that weak GFP signals could be interfered by background signals, whereas strong GFP signals could overshot the detection limitation of the flow cytometer and resulted in a deviation from linearity which was prevented by adjusting the setting in flow cytometer. Moreover, we compared promoter strength as measured by MFI × POI and found that the relative activity of CMV promoter to PGK promoter was 20 to 47 folds in A549 cells and 32 to > 100 folds in H1299 cells., Conclusions: The linear correlation between MFI × POI and a wide range of adenoviral MOI provides an efficient method to investigate factors regulating protein expression and to estimate virus titers.
- Published
- 2015
- Full Text
- View/download PDF
37. Transient receptor potential vanilloid type 1 is vital for (-)-epigallocatechin-3-gallate mediated activation of endothelial nitric oxide synthase.
- Author
-
Guo BC, Wei J, Su KH, Chiang AN, Zhao JF, Chen HY, Shyue SK, and Lee TS
- Subjects
- AMP-Activated Protein Kinases genetics, AMP-Activated Protein Kinases metabolism, Anilides pharmacology, Animals, Calcium metabolism, Capsaicin analogs & derivatives, Capsaicin pharmacology, Catechin pharmacology, Cell Movement drug effects, Cell Proliferation drug effects, Cell Survival drug effects, Cinnamates pharmacology, Endothelial Cells drug effects, HEK293 Cells, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Neovascularization, Pathologic metabolism, Nitric Oxide metabolism, Nitric Oxide Synthase Type III genetics, Phosphorylation, Signal Transduction, TRPV Cation Channels antagonists & inhibitors, TRPV Cation Channels genetics, Tea chemistry, Catechin analogs & derivatives, Nitric Oxide Synthase Type III metabolism, TRPV Cation Channels metabolism
- Abstract
Scope: Epigallocatechin-3-gallate (EGCG), the most abundant catechin of green tea, has beneficial effects on physiological functions of endothelial cells (ECs), yet the detailed mechanisms are not fully understood. In this study, we investigated the role of transient receptor potential vanilloid type 1 (TRPV1), a ligand-gated nonselective calcium channel, in EGCG-mediated endothelial nitric oxide (NO) synthase (eNOS) activation and angiogenesis., Methods and Results: In ECs, treatment with EGCG time-dependently increased the intracellular level of Ca(2+) . Removal of extracellular calcium (Ca(2+) ) by EGTA or EDTA or inhibition of TRPV1 by capsazepine or SB366791 abrogated EGCG-increased intracellular Ca(2+) level in ECs or TRPV1-transfected HEK293 cells. Additionally, EGCG increased the phsophorylation of eNOS at Ser635 and Ser1179, Akt at Ser473, calmodulin-dependent protein kinase II (CaMKII) at Thr286 and AMP-activated protein kinase (AMPK) at Thr172, all abolished by the TRPV1 antagonist capsazepine. EGCG-induced NO production was diminished by pretreatment with LY294002 (an Akt inhibitor), KN62 (a CaMKII inhibitor), and compound C (an AMPK inhibitor). Moreover, blocking TRPV1 activation prevented EGCG-induced EC proliferation, migration, and tube formation, as well as angiogenesis in Matrigel plugs in mice., Conclusion: EGCG may trigger activation of TRPV1-Ca(2+) signaling, which leads to phosphorylation of Akt, AMPK, and CaMKII; eNOS activation; NO production; and, ultimately, angiogenesis in ECs., (© 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2015
- Full Text
- View/download PDF
38. The essential role of transient receptor potential vanilloid 1 in simvastatin-induced activation of endothelial nitric oxide synthase and angiogenesis.
- Author
-
Su KH, Lin SJ, Wei J, Lee KI, Zhao JF, Shyue SK, and Lee TS
- Subjects
- Animals, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Mice, Inbred C57BL, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction physiology, Endothelial Cells metabolism, Neovascularization, Pathologic metabolism, Nitric Oxide Synthase Type III metabolism, Simvastatin pharmacology, TRPV Cation Channels metabolism
- Abstract
Aims: We investigated the role of transient receptor potential vanilloid receptor type 1 (TRPV1) in simvastatin-mediated activation of endothelial nitric oxide synthase (eNOS) and angiogenesis., Methods: Fluo-8 NW assay was for Ca(2+) detection; Griess's assay was for NO bioavailability; Western blotting and immunoprecipitation were for protein phosphorylation and interaction; tube formation and Matrigel plug assay were for angiogenesis., Results: In endothelial cells (ECs), treatment with simvastatin time-dependently increased intracellular level of Ca(2+). Pharmacological inhibition or genetic disruption of TRPV1 abrogated simvastatin-mediated elevation of intracellular Ca(2+) in ECs or TRPV1-transfected HEK293 cells. Loss of TRPV1 function abolished simvastatin-induced NO production and phosphorylation of eNOS and calmodulin protein kinase II (CaMKII) in ECs and in aortas of mice. Inhibition of TRPV1 activation prevented the simvastatin-elicited increase in the formation of TRPV1-Akt-CaMKII-AMPK-eNOS complex. In mice, Matrigel plug assay showed that simvastatin-evoked angiogenesis was abolished by TRPV1 antagonist and genetic ablation of TRPV1. Additionally, our results demonstrated that TRP ankyrin 1 (TRPA1) is the downstream effector in the simvastatin-activated TRPV1-Ca(2+) signalling and in the consequent NO production and angiogenesis as evidence by that re-expression of TRPA1 further augmented simvastatin-elicited Ca(2+) influx in TRPV1-expressed HEK293 cells and ablation of TRPA1 function profoundly inhibited the simvastatin-induced increase in the phosphorylation of eNOS and CaMKII, formation of TRPV1-Akt-CaMKII-AMPK-eNOS complex, NO bioavailability, tube formation and angiogenesis in ECs or mice., Conclusion: Simvastatin-induced Ca(2+) influx may through the activation of TRPV1-TRPA1 signalling, which leads to phosphorylation of CaMKII, increases in the formation of TRPV1-CaMKII-AMPK-eNOS complex, eNOS activation, NO production and, ultimately, angiogenesis in ECs., (© 2014 Scandinavian Physiological Society. Published by John Wiley & Sons Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
39. Implication of transient receptor potential vanilloid type 1 in 14,15-epoxyeicosatrienoic acid-induced angiogenesis.
- Author
-
Su KH, Lee KI, Shyue SK, Chen HY, Wei J, and Lee TS
- Subjects
- 8,11,14-Eicosatrienoic Acid pharmacology, Animals, Calcium metabolism, Gene Expression Regulation physiology, HEK293 Cells, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Nitric Oxide, Signal Transduction, TRPV Cation Channels genetics, 8,11,14-Eicosatrienoic Acid analogs & derivatives, Endothelial Cells physiology, Neovascularization, Physiologic drug effects, TRPV Cation Channels metabolism, Vasodilator Agents pharmacology
- Abstract
14,15-epoxyeicosatrienoic acid (14,15-EET) is implicated in regulating physiological functions of endothelial cells (ECs), yet the potential molecular mechanisms underlying the beneficial effects in ECs are not fully understood. In this study, we investigated whether transient receptor potential vanilloid receptor type 1 (TRPV1) is involved in 14,15-EET-mediated Ca(2+) influx, nitric oxide (NO) production and angiogenesis. In human microvascular endothelial cells (HMECs), 14,15-EET time-dependently increased the intracellular level of Ca(2+). Removal of extracellular Ca(2+), pharmacological inhibition or genetic disruption of TRPV1 abrogated 14,15-EET-mediated increase of intracellular Ca(2+) level in HMECs or TRPV1-transfected HEK293 cells. Furthermore, removal of extracellular Ca(2+) or pharmacological inhibition of TRPV1 decreased 14,15-EET-induced NO production. 14,15-EET-mediated tube formation was abolished by TRPV1 pharmacological inhibition. In an animal experiment, 14,15-EET-induced angiogenesis was diminished by inhibition of TRPV1 and in TRPV1-deficient mice. TRPV1 may play a crucial role in 14,15-EET-induced Ca(2+) influx, NO production and angiogenesis.
- Published
- 2014
- Full Text
- View/download PDF
40. 14-3-3σ induces heat shock protein 70 expression in hepatocellular carcinoma.
- Author
-
Liu CC, Jan YJ, Ko BS, Wu YM, Liang SM, Chen SC, Lee YM, Liu TA, Chang TC, Wang J, Shyue SK, Sung LY, and Liou JY
- Subjects
- Aged, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular pathology, Cell Movement genetics, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Female, Gene Expression Regulation, Neoplastic, HSP70 Heat-Shock Proteins metabolism, Heat Shock Transcription Factors, Hep G2 Cells, Humans, Liver Neoplasms metabolism, Liver Neoplasms pathology, Male, Middle Aged, Prognosis, Signal Transduction, Transcription Factors genetics, Transcription Factors metabolism, beta Catenin genetics, beta Catenin metabolism, 14-3-3 Proteins metabolism, Biomarkers, Tumor metabolism, Carcinoma, Hepatocellular genetics, Exoribonucleases metabolism, HSP70 Heat-Shock Proteins biosynthesis, Liver Neoplasms genetics
- Abstract
Background: 14-3-3σ is implicated in promoting tumor development of various malignancies. However, the clinical relevance of 14-3-3σ in hepatocellular carcinoma (HCC) tumor progression and modulation and pathway elucidation remain unclear., Methods: We investigated 14-3-3σ expression in 109 HCC tissues by immunohistochemistry. Overexpression and knockdown experiments were performed by transfection with cDNA or siRNA. Protein expression and cell migration were determined by Western blot and Boyden chamber assay., Results: In this study, we found that 14-3-3σ is abundantly expressed in HCC tumors. Stable or transient overexpression of 14-3-3σ induces the expression of heat shock factor-1α (HSF-1α) and heat shock protein 70 (HSP70) in HCC cells. Moreover, expression of 14-3-3σ significantly correlates with HSF-1α/HSP70 in HCC tumors and both 14-3-3σ and HSP70 overexpression are associated with micro-vascular thrombi in HCC patients, suggesting that 14-3-3σ/HSP70 expression is potentially involved in cell migration/invasion. Results of an in vitro migration assay indicate that 14-3-3σ promotes cell migration and that 14-3-3σ-induced cell migration is impaired by siRNA knockdown of HSP70. Finally, 14-3-3σ-induced HSF-1α/HSP70 expression is abolished by the knockdown of β-catenin or activation of GSK-3β., Conclusions: Our findings indicate that 14-3-3σ participates in promoting HCC cell migration and tumor development via β-catenin/HSF-1α/HSP70 pathway regulation. Thus, 14-3-3σ alone or combined with HSP70 are potential prognostic biomarkers for HCC.
- Published
- 2014
- Full Text
- View/download PDF
41. Activation of soluble guanylyl cyclase prevents foam cell formation and atherosclerosis.
- Author
-
Tsou CY, Chen CY, Zhao JF, Su KH, Lee HT, Lin SJ, Shyue SK, Hsiao SH, and Lee TS
- Subjects
- ATP Binding Cassette Transporter 1 genetics, ATP Binding Cassette Transporter 1 metabolism, Animals, Apolipoproteins E genetics, Apolipoproteins E metabolism, Cell Line, Foam Cells cytology, Guanylate Cyclase genetics, Indazoles pharmacology, Lipid Metabolism, Lipoproteins, LDL metabolism, Liver enzymology, Liver X Receptors, Macrophages enzymology, Macrophages metabolism, Mice, Mice, Knockout, Myocardium enzymology, Orphan Nuclear Receptors genetics, Orphan Nuclear Receptors metabolism, Atherosclerosis metabolism, Enzyme Activation physiology, Foam Cells physiology, Guanylate Cyclase metabolism
- Abstract
Aims: Soluble guanylyl cyclase (sGC) is a key modulator in the regulation of vascular tone. However, its role and involving mechanism in cholesterol metabolism of macrophages and atherosclerosis remain unclear., Methods: Oil red O staining, Dil-oxidized low-density lipoprotein (oxLDL)-binding assay and cholesterol efflux assay were performed in biology of foam cells. Levels of cytokines or intracellular lipid were evaluated by ELISA or colorimetric kits. Expression of gene or protein was determined by quantitative real-time PCR or Western blotting. Histopathology was examined by haematoxylin and eosin staining., Results: Soluble guanylyl cyclase was expressed in macrophages of mouse atherosclerotic lesions. Treatment with 1H-[1, 2, 4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ, sGC inhibitor) exacerbated oxLDL-induced cholesterol accumulation in macrophages. In contrast, 3-(5'-hydroxymethyl-2'furyl)-1-benzyl indazole (YC-1, sGC activator) attenuated the oxLDL-induced cholesterol accumulation because of increased cholesterol efflux. Additionally, YC-1 dose dependently increased the protein expression of ATP-binding cassette transporter A1 (ABCA1) but did not alter that of scavenger receptor class A (SR-A), CD36, SR-BI or ABCG1. Moreover, YC-1-upregulated ABCA1 level depended on liver X receptor α (LXRα). Inhibition of the LXRα-ABCA1 pathway by LXRα small interfering RNA (siRNA), ABCA1 neutralizing antibody or ABCA1 siRNA abolished the effect of YC-1 on cholesterol accumulation and cholesterol efflux. In vivo, YC-1 retarded the development of atherosclerosis, accompanied by reduced serum levels of cholesterol and pro-inflammatory cytokines, in apolipoprotein E-deficient mice., Conclusion: Activation of sGC by YC-1 leads to LXRα-dependent upregulation of ABCA1 in macrophages and may confer protection against atherosclerosis., (© 2013 Scandinavian Physiological Society. Published by John Wiley & Sons Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
42. Role of glycine N-methyltransferase in experimental ulcerative colitis.
- Author
-
Chou WY, Zhao JF, Chen YM, Lee KI, Su KH, Shyue SK, and Lee TS
- Subjects
- Animals, Colitis, Ulcerative chemically induced, Colitis, Ulcerative pathology, Dextran Sulfate, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, Gene Expression, Glycine N-Methyltransferase metabolism, Inflammation Mediators metabolism, Male, Mice, Mice, Inbred C57BL, Molecular Targeted Therapy, Colitis, Ulcerative genetics, Glycine N-Methyltransferase genetics, Glycine N-Methyltransferase physiology
- Abstract
Background and Aim: Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders with unclear etiology and mechanism(s). Glycine N-methyltransferase (GNMT) plays a central role in inflammatory diseases such as hepatitis and atherosclerosis. However, little is known about the impact of GNMT and the involved mechanism in the pathogenesis of IBD. In the current study, we investigated the role of GNMT in the mouse model of dextran sulfate sodium (DSS)-induced colitis., Methods: Protein expression was determined by Western blotting or immunohistochemistry. Histopathology was examined by hematoxylin and eosin staining. Levels of pro-inflammatory cytokines were evaluated by ELISA kits., Results: GNMT was expressed in the epithelium of the colon under normal conditions, and with DSS treatment, its expression was predominant in infiltrated leukocytes of lesions. Mice with genetic deletion of GNMT (GNMT(-/-) ) showed increased susceptibility to DSS induction of colitis, as revealed by the progression of colitis. Additionally, severe colonic inflammation, including increased crypt loss, leukocyte infiltration, and hemorrhage, was greater with DSS treatment in GNMT(-/-) than wild-type mice. Furthermore, the expression of adhesion molecule and inflammatory mediators in the colon was significantly higher with DSS treatment in GNMT(-/-) than wild-type mice. Moreover, loss of GNMT decreased cell apoptosis in colitis lesions with DSS treatment., Conclusions: Collectively, our findings suggest that GNMT may be a crucial molecule in the pathogenesis of DSS-induced colitis. This finding may provide new information for a potential therapeutic target in treating IBD., (© 2013 Journal of Gastroenterology and Hepatology Foundation and Wiley Publishing Asia Pty Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
43. Excess nitric oxide impairs LXR(α)-ABCA1-dependent cholesterol efflux in macrophage foam cells.
- Author
-
Zhao JF, Shyue SK, Lin SJ, Wei J, and Lee TS
- Subjects
- ATP Binding Cassette Transporter 1 biosynthesis, Animals, Apolipoproteins E genetics, Atherosclerosis chemically induced, Foam Cells drug effects, Foam Cells metabolism, Gene Expression Regulation drug effects, Lipoproteins, LDL, Liver drug effects, Liver metabolism, Liver X Receptors, Macrophages drug effects, Mice, Nitric Oxide toxicity, Nitric Oxide Synthase Type II metabolism, Orphan Nuclear Receptors biosynthesis, Oxidation-Reduction, S-Nitroso-N-Acetylpenicillamine administration & dosage, ATP Binding Cassette Transporter 1 metabolism, Atherosclerosis metabolism, Lipid Metabolism drug effects, Nitric Oxide metabolism, Orphan Nuclear Receptors metabolism
- Abstract
Excess nitric oxide (NO) promotes the progression of atherosclerosis by increasing the oxidation of low-density lipoprotein (LDL) and inflammatory responses. However, little is known about the impact of NO and its underlying molecular mechanism on lipid metabolism of macrophage foam cells. In this study, Oil-red O staining, cholesterol and triglyceride assay, Dil-oxidized LDL (oxLDL) binding assay, cholesterol efflux assay, real-time RT-PCR and Western blot analysis were used for in vitro experiments. Apolipoprotein E-deficient (apoE(-/-) ) and apoE and inducible nitric oxide synthase-deficient (apoE(-/-) iNOS(-/-) ) mice were as our in vivo models. Treatment with S-nitroso-N-acetyl-D,L-penicillamine (SNAP), an NO donor, exacerbated oxLDL-induced cholesterol accumulation in macrophages, because of reduced efficacy of cholesterol efflux. In addition, SNAP decreased the protein level of ATP-binding cassette transporter A1 (ABCA1) without affecting scavenger receptor type A (SR-A), CD36, ABCG1, or SR-B1 levels. This SNAP-mediated downregulation of ABCA1 was mainly through the effect of NO but not peroxynitrite. Furthermore, the SNAP-downregulated ABCA1 was due to the decrease in the liver X receptor α (LXRα)-dependent transcriptional regulation. Moreover, genetic deletion of iNOS increased the serum capacity of reverse cholesterol efflux and protein expression of LXRα, ABCA1, and SR-BI in aortas and retarded atherosclerosis in apoE(-/-) mice. Our findings provide new insights in the pro-atherogenic effect of excess NO on cholesterol metabolism in macrophages., (© 2013 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
44. Caveolin-1 interacts with Derlin-1 and promotes ubiquitination and degradation of cyclooxygenase-2 via collaboration with p97 complex.
- Author
-
Chen SF, Wu CH, Lee YM, Tam K, Tsai YC, Liou JY, and Shyue SK
- Subjects
- Adaptor Proteins, Vesicular Transport, Adenosine Triphosphatases genetics, Animals, Caveolin 1 genetics, Cell Cycle Proteins genetics, Cyclooxygenase 2 genetics, HEK293 Cells, Humans, Intercellular Signaling Peptides and Proteins, Intracellular Signaling Peptides and Proteins, Membrane Proteins genetics, Mice, Multiprotein Complexes genetics, Protein Transport physiology, Proteins genetics, Proteins metabolism, Valosin Containing Protein, Adenosine Triphosphatases metabolism, Caveolin 1 metabolism, Cell Cycle Proteins metabolism, Cyclooxygenase 2 metabolism, Membrane Proteins metabolism, Multiprotein Complexes metabolism, Proteolysis, Ubiquitination physiology
- Abstract
Caveolin-1 (Cav-1) interacts with and mediates protein trafficking and various cellular functions. Derlin-1 is a candidate for the retrotranslocation channel of endoplasmic reticulum proteins. However, little is known about how Derlin-1 mediates glycosylated protein degradation. Here, we identified Cav-1 as a key player in Derlin-1- and p97-mediated cyclooxygenase 2 (COX-2) ubiquitination and degradation. Derlin-1 augmented the interaction of Cav-1 and COX-2 and mediated the degradation of COX-2 in a COX-2 C terminus-dependent manner. Suppression of Cav-1 decreased the ubiquitination of COX-2, and mutation of Asn-594 to Ala to disrupt N-glycosylation at the C terminus of COX-2 reduced the interaction of COX-2 with Cav-1 but not Derlin-1. Moreover, suppression of p97 increased the ubiquitination of COX-2 and up-regulated COX-2 but not COX-1. Cav-1 enhanced the interaction of p97 with Ufd1 and Derlin-1 and collaborated with p97 to interact with COX-2. Cav-1 may be a cofactor in the interaction of Derlin-1 and N-glycosylated COX-2 and may facilitate Derlin-1- and p97 complex-mediated COX-2 ubiquitination, retrotranslocation, and degradation.
- Published
- 2013
- Full Text
- View/download PDF
45. Activation of transient receptor potential vanilloid 1 decreases endothelial nitric oxide synthase phosphorylation at Thr497 by protein phosphatase 2B-dependent dephosphorylation of protein kinase C.
- Author
-
Ching LC, Zhao JF, Su KH, Shyue SK, Hsu CP, Lu TM, Lin SJ, and Lee TS
- Subjects
- Animals, Blotting, Western, Cattle, Immunoprecipitation, Mice, Mice, Knockout, Phosphorylation, Signal Transduction physiology, Calcineurin metabolism, Endothelial Cells metabolism, Nitric Oxide Synthase Type III metabolism, Protein Kinase C metabolism, TRPV Cation Channels metabolism
- Abstract
Aims: We investigated the effects and underlying molecular mechanism of transient receptor potential vanilloid 1 (TRPV1), a calcium (Ca(2+) )-permeable non-selective cation channel, on phosphorylation of endothelial nitric oxide synthase (eNOS) at threonine 497 (Thr497) in bovine aortic endothelial cells (BAECs) and in mice., Methods: Western blotting and immunoprecipitation were used for the evaluation of protein phosphorylation; protein phosphatase 2B (PP2B) activity was assessed by convention kit; Griess assay was for NO production; tube formation and Matrigel plug assay were used for angiogenesis., Results: In BAECs, treatment with the TRPV1 ligand evodiamine decreased the phosphorylation of eNOS at Thr497, protein kinase Cα (PKCα) at Serine 657 (Ser657) and PKCβ2 at Ser660. Evodiamine increased protein phosphatase 2B (PP2B) activity and promoted the formation of a PP2B-PKC complex. Inhibition of TRPV1 activation by the pharmacological antagonists, removal of extracellular Ca(2+) or pharmacological inhibition of PI3K/Akt/calmodulin-dependent protein kinase II/AMP-activated protein kinase signalling pathway abolished the evodiamine-induced alterations in phosphorylation of eNOS at Thr497, PKCα at Ser657, PKCβ2 at Ser660 and PP2B activity, as well as the formation of a PP2B-PKC complex. Inhibition of PP2B activation partially reduced the evodiamine-induced NO bioavailability and tube formation in endothelial cells (ECs) and angiogenesis in mice. Moreover, evodiamine decreased the phosphorylation of eNOS at Thr497, PKCα at Ser657 and PKCβ2 at Ser660 in apolipoprotein E (ApoE)-deficient mouse aortas but not TRPV1-deficient or ApoE/TRPV1 double-knockout mice., Conclusion: TRPV1 activation in ECs may elicit a Ca(2+) -dependent effect on PP2B-PKC signalling, which leads to dephosphorylation of eNOS at Thr497 in ECs and in mice., (© 2013 Scandinavian Physiological Society. Published by John Wiley & Sons Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
46. Essential role of transient receptor potential vanilloid type 1 in evodiamine-mediated protection against atherosclerosis.
- Author
-
Wei J, Ching LC, Zhao JF, Shyue SK, Lee HF, Kou YR, and Lee TS
- Subjects
- Animals, Apolipoproteins E deficiency, Blotting, Western, Coronary Artery Disease genetics, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, Fatty Liver metabolism, Fatty Liver pathology, Hyperlipidemias metabolism, Hyperlipidemias pathology, Male, Mice, Mice, Knockout, TRPV Cation Channels genetics, Coronary Artery Disease metabolism, Coronary Artery Disease pathology, Quinazolines pharmacology, TRPV Cation Channels metabolism
- Abstract
Aim: We investigated whether transient receptor potential vanilloid type 1 (TRPV1) was involved in the therapeutic effect of evodiamine, a main bioactive component in the fruit of Evodiae rutaecarpa, on the development of atherosclerosis in apolipoprotein E-deficient (ApoE(-/-)) mice and ApoE(-/-)TRPV1(-/-) mice., Methods: Histopathology was examined by haematoxylin and eosin staining, levels of cytokines and mediators were evaluated by ELISA kits, and protein expression was determined by Western blotting., Results: Chronic administration with evodiamine (10 mg kg(-1) body weight) reduced the size of atherosclerotic lesions and alleviated the hyperlipidaemia and systemic inflammation, as well as hepatic macrovesicular steatosis, in ApoE(-/-) mice. Treating ApoE(-/-) mice with evodiamine enhanced hepatic cholesterol clearance, as revealed by upregulation of hepatic low-density lipoprotein receptor and ATP-binding cassette (ABC) transporters ABCG5, ABCG8 and cholesterol 7α-hydrolase. Genetic deletion of TRPV1 in ApoE(-/-) mice promoted the progression of atherosclerosis; elevated the serum levels of cholesterol, cytokines and chemokines; and exacerbated hepatic macrovesicular steatosis. Moreover, genetic deletion of TRPV1 abrogated the evodiamine-evoked atheroprotection but not anti-obesity effect in ApoE(-/-) mice., Conclusion: Evodiamine may confer novel TRPV1-dependent atheroprotection and TRPV1-independent anti-obesity action., (© 2012 The Authors Acta Physiologica © 2012 Scandinavian Physiological Society.)
- Published
- 2013
- Full Text
- View/download PDF
47. Novel effect of paeonol on the formation of foam cells: promotion of LXRα-ABCA1-dependent cholesterol efflux in macrophages.
- Author
-
Zhao JF, Jim Leu SJ, Shyue SK, Su KH, Wei J, and Lee TS
- Subjects
- ATP Binding Cassette Transporter 1 genetics, ATP Binding Cassette Transporter 1 physiology, Acetophenones therapeutic use, Animals, Atherosclerosis drug therapy, Atherosclerosis etiology, Cell Culture Techniques, Dose-Response Relationship, Drug, Inflammation Mediators metabolism, Lipoproteins, LDL metabolism, Liver X Receptors, Male, Mice, Molecular Targeted Therapy, Orphan Nuclear Receptors genetics, Orphan Nuclear Receptors physiology, Phytotherapy, RNA, Small Interfering, ATP Binding Cassette Transporter 1 metabolism, Acetophenones pharmacology, Cholesterol metabolism, Foam Cells metabolism, Gene Expression drug effects, Orphan Nuclear Receptors metabolism, Up-Regulation drug effects
- Abstract
Paeonol, a phenolic component purified from Paeonia suffruticosa (Cortex Moutan), is used in traditional Chinese medicine to treat inflammatory diseases. However, little is known about the effect of paeonol on cholesterol metabolism. We investigated the efficacy of paeonol on cholesterol metabolism and the underlying mechanism in macrophages and apolipoprotein E deficient (apoE(-/-)) mice. Treatment with paeonol markedly attenuated cholesterol accumulation induced by oxidized LDL in macrophages, which was due to increased cholesterol efflux. Additionally, paeonol enhanced the mRNA and protein expression of ATP-binding membrane cassette transport protein A1 (ABCA1) but did not alter the protein level of ABCG1 or other scavenger receptors. Inhibition of ABCA1 activity with a pharmacological inhibitor, neutralizing antibody or small interfering RNA (siRNA), negated the effects of paeonol on cholesterol efflux and cholesterol accumulation. Furthermore, paeonol induced the nuclear translocation of liver X receptor α (LXRα) by increasing its activity. siRNA knockdown of LXRα abolished the paeonol-induced upregulation of ABCA1, promotion of cholesterol efflux and suppression of cholesterol accumulation. Moreover, atherosclerotic lesions, hyperlipidemia and systemic inflammation were reduced and the protein expression of ABCA1 was increased in aortas of paeonol-treated apoE(-/-) mice. Paeonol may alleviate the formation of foam cells by enhancing LXRα-ABCA1-dependent cholesterol efflux.
- Published
- 2013
- Full Text
- View/download PDF
48. Activation of TRPV1 prevents OxLDL-induced lipid accumulation and TNF-α-induced inflammation in macrophages: role of liver X receptor α.
- Author
-
Zhao JF, Ching LC, Kou YR, Lin SJ, Wei J, Shyue SK, and Lee TS
- Subjects
- Animals, Atherosclerosis genetics, Atherosclerosis metabolism, Blotting, Western, Calcium metabolism, Capsaicin analogs & derivatives, Capsaicin pharmacology, Cells, Cultured, Chromatin Immunoprecipitation, Inflammation chemically induced, Liver X Receptors, Macrophages drug effects, Mice, Orphan Nuclear Receptors genetics, Orphan Nuclear Receptors metabolism, RNA, Small Interfering, TRPV Cation Channels agonists, TRPV Cation Channels antagonists & inhibitors, TRPV Cation Channels genetics, Tumor Necrosis Factor-alpha pharmacology, Inflammation metabolism, Lipoproteins, LDL pharmacology, Macrophages metabolism, TRPV Cation Channels metabolism
- Abstract
The transient receptor potential vanilloid type 1 (TRPV1) is crucial in the pathogenesis of atherosclerosis; yet its role and underlying mechanism in the formation of macrophage foam cells remain unclear. Here, we show increased TRPV1 expression in the area of foamy macrophages in atherosclerotic aortas of apolipoprotein E-deficient mice. Exposure of mouse bone-marrow-derived macrophages to oxidized low-density lipoprotein (oxLDL) upregulated the expression of TRPV1. In addition, oxLDL activated TRPV1 and elicited calcium (Ca(2+)) influx, which were abrogated by the pharmacological TRPV1 antagonist capsazepine. Furthermore, oxLDL-induced lipid accumulation in macrophages was ameliorated by TRPV1 agonists but exacerbated by TRPV1 antagonist. Treatment with TRPV1 agonists did not affect the internalization of oxLDL but promoted cholesterol efflux by upregulating the efflux ATP-binding cassette (ABC) transporters ABCA1 and ABCG1. Moreover, the upregulation of ABC transporters was mainly through liver X receptor α-(LXRα-) dependent regulation of transcription. Moreover, the TNF-α-induced inflammatory response was alleviated by TRPV1 agonists but aggravated by the TRPV1 antagonist and LXR α siRNA in macrophages. Our data suggest that LXR α plays a pivotal role in TRPV1-activation-conferred protection against oxLDL-induced lipid accumulation and TNF-α-induced inflammation in macrophages.
- Published
- 2013
- Full Text
- View/download PDF
49. Enhanced prostacyclin synthesis by adenoviral gene transfer reduced glial activation and ameliorated dopaminergic dysfunction in hemiparkinsonian rats.
- Author
-
Tsai MJ, Weng CF, Yu NC, Liou DY, Kuo FS, Huang MC, Huang WC, Tam K, Shyue SK, and Cheng H
- Subjects
- Animals, Benzophenones pharmacology, Carbon Isotopes, Cell Proliferation drug effects, Cells, Cultured, Cyclooxygenase 2 metabolism, Cytochrome P-450 Enzyme System metabolism, Dopaminergic Neurons drug effects, Dopaminergic Neurons metabolism, Green Fluorescent Proteins metabolism, Imidazoles pharmacology, Intramolecular Oxidoreductases metabolism, Lipopolysaccharides pharmacology, Mesencephalon pathology, Neuroglia drug effects, Neuroglia pathology, Nitric Oxide biosynthesis, Nitric Oxide Synthase Type II metabolism, Oxidopamine, Rats, Rats, Sprague-Dawley, Substantia Nigra drug effects, Substantia Nigra metabolism, Substantia Nigra pathology, Transduction, Genetic, Adenoviridae metabolism, Dopaminergic Neurons pathology, Epoprostenol biosynthesis, Gene Transfer Techniques, Neuroglia metabolism, Parkinson Disease metabolism, Parkinson Disease pathology
- Abstract
Prostacyclin (PGI2), a potent vasodilator and platelet antiaggregatory eicosanoid, is cytoprotective in cerebral circulation. It is synthesized from arachidonic acid (AA) by the sequential action of cyclooxygenase- (COX-) 1 or 2 and prostacyclin synthase (PGIS). Because prostacyclin is unstable in vivo, PGI2 analogs have been developed and demonstrated to protect against brain ischemia. This work attempts to selectively augment PGI2 synthesis in mixed glial culture or in a model of Parkinson's disease (PD) by direct adenoviral gene transfer of prostacyclin biosynthetic enzymes and examines whether it confers protection in cultures or in vivo. Confluent mixed glial cultures actively metabolized exogenous AA into PGE2 and PGD2. These PGs were largely NS398 sensitive and considered as COX-2 products. Gene transfer of AdPGIS to the cultures effectively shunted the AA catabolism to prostacyclin synthesis and concurrently reduced cell proliferation. Furthermore, PGIS overexpression significantly reduced LPS stimulation in cultures. In vivo, adenoviral gene transfer of bicistronic COX-1/PGIS to substantia nigra protected 6-OHDA- induced dopamine depletion and ameliorated behavioral deficits. Taken together, this study shows that enhanced prostacyclin synthesis reduced glial activation and ameliorated motor dysfunction in hemiparkinsonian rats. Prostacyclin may have a neuroprotective role in modulating the inflammatory response in degenerating nigra-striatal pathway.
- Published
- 2013
- Full Text
- View/download PDF
50. Inflammatory role of AMP-activated protein kinase signaling in an experimental model of toxic smoke inhalation injury.
- Author
-
Perng DW, Chang TM, Wang JY, Lee CC, Lu SH, Shyue SK, Lee TS, and Kou YR
- Subjects
- Animals, Cells, Cultured, Humans, Inflammation enzymology, Inflammation immunology, Inflammation pathology, Male, Mice, Mice, Inbred C57BL, NADPH Oxidases metabolism, Random Allocation, Reactive Oxygen Species metabolism, Respiratory Mucosa immunology, Smoke Inhalation Injury pathology, AMP-Activated Protein Kinases metabolism, Interleukin-8 metabolism, MAP Kinase Signaling System, Smoke Inhalation Injury enzymology, Smoke Inhalation Injury immunology
- Abstract
Objective: The molecular mechanisms underlying lung inflammation in toxic smoke inhalation injury are unknown. We investigated the signaling pathway responsible for the induction of interleukin 8 by wood smoke extract in lung epithelial cells and lung inflammation induced by wood smoke exposure in mice., Design: A randomized, controlled study., Setting: A research laboratory., Interventions and Main Results: Exposure of primary human bronchial epithelial cells to wood smoke extract sequentially activated NADPH oxidase and increased intracellular reactive oxygen species level; activated AMP-activated protein kinase, extracellular signal-regulated kinase and Jun N-terminal kinase (two mitogen-activated protein kinases), and nuclear factor-κB and signal transducer and activator of transcription protein 3 (two transcription factors); and induced interleukin-8. Inhibition of NADPH oxidase activation with apocynin or siRNA targeting p47(phox ) (a subunit of NADPH oxidase) attenuated the increased intracellular reactive oxygen species level, AMP-activated protein kinase activation, and interleukin-8 induction. Removal of intracellular reactive oxygen species by N-acetyl-cysteine reduced the activation of AMP-activated protein kinase, extracellular signal-regulated kinase and Jun N-terminal kinase, and interleukin-8 induction. Prevention of AMP-activated protein kinase activation by Compound C or AMP-activated protein kinase siRNA lessened the activation of Jun N-terminal kinase, extracellular signal-regulated kinase, nuclear factor-κB, signal transducer and activator of transcription protein 3 and interleukin-8 induction. Inhibition of Jun N-terminal kinase and extracellular signal-regulated kinase activation by inhibitors reduced the activation of nuclear factor-κB and signal transducer and activator of transcription protein 3 and interleukin-8 induction. Abrogation of nuclear factor-κB and signal transducer and activator of transcription protein 3 activation by inhibitors attenuated the interleukin-8 induction. Additionally, acute exposure of mice to wood smoke promoted AMP-activated protein kinase phosphorylation and expression of macrophage inflammatory protein 2 (an interleukin-8 homolog) in lung epithelial cells and lungs and lung inflammation, all of which were reduced by Compound C treatment., Conclusions: Interleukin-8 induction by wood smoke extract in lung epithelial cells is mediated by novel NADPH oxidase-dependent, reactive oxygen species-sensitive AMP-activated protein kinase signaling with Jun N-terminal kinase and extracellular signal-regulated kinase as the downstream kinases and nuclear factor-κB and signal transducer and activator of transcription protein 3 as the downstream transcription factors. This AMP-activated protein kinase signaling is likely important for inducing lung inflammation with toxic smoke exposure in mice.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.