Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Arch Toxicol ; 2024 May 16.
Article in English | MEDLINE | ID: mdl-38753188

ABSTRACT

Long interspersed nuclear element-1 (LINE-1) methylation serves as an indicator of global DNA methylation. This study explored the correlation between LINE-1 methylation and chronic kidney disease (CKD). We also evaluated whether LINE-1 methylation could modify the association between CKD and metal exposure. A total of 213 patients with clinically defined CKD, without hemodialysis and 416 age and sex matched controls were recruited. Levels of LINE-1 methylation, total urinary arsenic, blood lead, blood cadmium, and plasma selenium were assessed. The results reveal a positive association between LINE-1 methylation and CKD, with an odds ratio (OR) of 5.30 (95% confidence interval: 2.81 to 9.99). Total urinary arsenic and blood cadmium concentrations were positively related with LINE-1 methylation. This study was the first to observe that low plasma selenium, high blood cadmium, and high blood lead levels significantly and additively interact with increased LINE-1 methylation to increase the OR of CKD. Additionally, high LINE-1 methylation interacted multiplicatively with low plasma selenium to increase the OR of CKD (p < 0.001). This study highlighted the significant association between LINE-1 hypermethylation and CKD. Furthermore, the results demonstrate that LINE-1 methylation can interact with high blood cadmium or low plasma selenium to affect CKD risk.

2.
Int J Mol Sci ; 24(9)2023 May 01.
Article in English | MEDLINE | ID: mdl-37175838

ABSTRACT

Adiponectin is an adipokine multipeptide hormone with insulin-sensitizing; anti-atherosclerotic; and anti-inflammatory properties. Chronic kidney disease (CKD) may be associated with low adiponectin. The adiponectin gene ADIPOQ is thought to be the only major gene responsible for plasma adiponectin levels; which are associated with diabetes and diabetic nephropathy. The purpose of this study was to investigate the association between ADIPOQ polymorphism and CKD. In addition; the combined effects of ADIPOQ polymorphism and diabetes and levels of total urinary arsenic and blood cadmium on CKD were also explored. This study included 215 CKD patients and 423 age-sex matched controls. The ADIPOQ polymorphisms were determined using the Agena Bioscience Mass ARRAY System. The levels of blood cadmium and urinary arsenic species were measured. The ADIPOQ rs182052 GA/AA genotype had a marginally lower odds ratio (OR) for CKD than the GG genotype. The OR (95% confidence interval; CI) was 16.33 (5.72-46.66) of CKD in subjects carrying the ADIPOQ rs182052 GG genotype and diabetes compared to non-diabetes subjects carrying the ADIPOQ rs182052 GA/AA genotype; the interaction term had p = 0.015; and the synergy index was 6.64 (1.81-24.36) after multivariate adjustment. A significant interaction of diabetes and ADIPOQ rs1501299 risk genotype increased the OR of CKD after multivariate adjustment with a synergy index of 0.31 (0.11-0.86) and a multiplicative interaction with p = 0.001. These results suggest that ADIPOQ rs182052 and rs1501299 risk genotypes may significantly modify the association between diabetes and CKD but not the association between total urinary arsenic and blood cadmium and CKD.


Subject(s)
Adiponectin , Arsenic , Diabetes Mellitus , Diabetic Nephropathies , Humans , Adiponectin/genetics , Cadmium , Case-Control Studies , Diabetes Mellitus/genetics , Diabetic Nephropathies/genetics , Genetic Predisposition to Disease , Genotype , Polymorphism, Single Nucleotide
3.
Biochem Biophys Res Commun ; 650: 73-80, 2023 04 02.
Article in English | MEDLINE | ID: mdl-36773342

ABSTRACT

Intracellular lipid droplet (LD) generation is the primary site of energy storage, which is necessary for physiological homeostasis but is related to pathological metabolic disorders. Lipid metabolism is critical for maintaining innate and adaptive immunity; however, it is mainly undefined in peripheral immune cells. Flow cytometry-based immune profiling in healthy peripheral blood cells showed significant original generation of LDs in dendritic cells (DCs, CD3-CD19-CD56-CD11+), monocytes (CD3-CD19-CD56-CD14+), natural killer cells (NK, CD3-CD19-CD56+), and B cells (CD3-CD19+). CD36, a common scavenger receptor of lipids, was also highly expressed in LD-accumulated DCs and monocytes. Following short-term treatment with oxidized LDL (oxLDL) in an experimental ex vivo model, CD14+ monocytes showed an effective increase in LD generation, but there were no alterations in the immune cell populations. Furthermore, oxLDL-treated CD14+ monocytes displayed CD36 expression. However, oxLDL-primed CD14+ monocytes showed a blockade in the uptake of extra oxLDL, even while expressing increased CD36, indicating a defect in lipid clearance. Exogenous treatment with oxLDL caused monocyte type 1 polarization accompanied by increased LD accumulation and CD36 expression. This study describes a method to monitor LD generation and CD36 expression in peripheral immune cells and identified an immunomodulatory effect of oxLDL on monocytes by tilting them towards type 1 polarization.


Subject(s)
Dyslipidemias , Monocytes , Humans , Monocytes/metabolism , Lipid Droplets/metabolism , Lipoproteins, LDL/metabolism , Receptors, Scavenger/metabolism , CD36 Antigens/metabolism , Dyslipidemias/metabolism
4.
Breast Cancer Res Treat ; 189(3): 665-676, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34553296

ABSTRACT

PURPOSE: Neratinib, an irreversible pan-HER tyrosine kinase inhibitor, has demonstrated systemic efficacy and intracranial activity in various stages of HER2+breast cancer. NALA was a phase III randomized trial that assessed the efficacy and safety of neratinib+capecitabine (N+C) against lapatinib+capecitabine (L+C) in HER2+ metastatic breast cancer (mBC) patients who had received ≥ 2 HER2-directed regimens. Descriptive analysis results of the Asian subgroup in the NALA study are reported herein. METHODS: 621 centrally assessed HER2+ mBC patients were enrolled, 202 of whom were Asian. Those with stable, asymptomatic brain metastases (BM) were eligible for study entry. Patients were randomized 1:1 to N (240 mg qd) + C (750 mg/m2 bid, day 1-14) with loperamide prophylaxis or to L (1250 mg qd) + C (1000 mg/m2 bid, day 1-14) in 21-day cycles. Co-primary endpoints were centrally assessed progression-free survival (PFS) and overall survival (OS). Secondary endpoints included time to intervention for central nervous system (CNS) disease, objective response rate, duration of response (DoR), clinical benefit rate, and safety. RESULTS: 104 and 98 Asian patients were randomly assigned to receive N+C or L+C, respectively. Median PFS of N+C and L+C was 7.0 and 5.4 months (P = 0.0011), respectively. Overall cumulative incidence of intervention for CNS disease was lower with N+C (27.9 versus 33.8%; P = 0.039). Both median OS (23.8 versus 18.7 months; P = 0.185) and DoR (11.1 versus 4.2 months; P < 0.0001) were extended with N+C, compared to L+C. The incidences of grade 3/4 treatment emergent adverse events (TEAEs) and TEAEs leading to treatment discontinuation were mostly comparable between the two arms. Diarrhea and palmar-plantar erythrodysesthesia were the most frequent TEAEs in both arms, similar to the overall population in incidence and severity. CONCLUSION: Consistent with the efficacy profile observed in the overall study population, Asian patients with HER2+ mBC, who had received ≥ 2 HER2-directed regimens, may also benefit from N+C. No new safety signals were noted. CLINICAL TRIAL REGISTRATION: NCT01808573.


Subject(s)
Breast Neoplasms , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Capecitabine/therapeutic use , Female , Humans , Lapatinib/therapeutic use , Quinolines , Receptor, ErbB-2/genetics , Treatment Outcome
5.
Int J Mol Sci ; 18(12)2017 Dec 13.
Article in English | MEDLINE | ID: mdl-29236027

ABSTRACT

Malignant human anaplastic thyroid cancer (ATC) is pertinacious to conventional therapies. The present study investigated the anti-cancer activity of simvastatin and its underlying regulatory mechanism in cultured ATC cells. Simvastatin (0-20 µM) concentration-dependently reduced cell viability and relative colony formation. Depletions of mevalonate (MEV) and geranylgeranyl pyrophosphate (GGpp) by simvastatin induced G1 arrest and increased apoptotic cell populations at the sub-G1 phase. Adding MEV and GGpp prevented the simvastatin-inhibited cell proliferation. Immunoblotting analysis illustrated that simvastatin diminished the activation of RhoA and Rac1 protein, and this effect was prevented by pre-treatment with MEV and GGpp. Simvastatin increased the levels of p21cip and p27kip proteins and reduced the levels of hyperphosphorylated-Rb, E2F1 and CCND1 proteins. Adding GGpp abolished the simvastatin-increased levels of p27kip protein, and the GGpp-caused effect was abolished by Skp2 inhibition. Introduction of Cyr61 siRNA into ATC cells prevented the epidermal growth factor (EGF)-enhanced cell migration. The EGF-induced increases of Cyr61 protein expression and cell migration were prevented by simvastatin. Taken together, these results suggest that simvastatin induced ATC proliferation inhibition through the deactivation of RhoA/Rac1 protein and overexpression of p21cip and p27kip, and migration inhibition through the abrogation of Cyr61 protein expression.


Subject(s)
Cell Proliferation/drug effects , Simvastatin/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cysteine-Rich Protein 61/antagonists & inhibitors , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Epidermal Growth Factor/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Mevalonic Acid/pharmacology , Polyisoprenyl Phosphates/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Simvastatin/therapeutic use , Thyroid Carcinoma, Anaplastic/drug therapy , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
6.
Immunol Res ; 64(2): 369-84, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26071024

ABSTRACT

Connective tissue growth factor (CTGF), a CCN family member, is a secreted protein regulating cellular functions, including fibrosis, apoptosis, adhesion, migration, differentiation, proliferation, angiogenesis, and chondrogenesis. CTGF increases proinflammatory factor production; however, inflammatory cytokine regulation by CTGF is poorly understood. The aim of this study was to identify novel biological functions and elucidate the functional mechanisms of CTGF. Specifically, the study focused on the ability of CTGF-primed monocytes to secrete interleukin 8 (CXCL8/IL-8) and determined the signaling pathways involved in CTGF-induced CXCL8/IL-8 gene regulation during inflammation. We transfected wild-type or mutant CXCL8/IL-8 promoter-derived luciferase reporter constructs into 293T cells to examine the effect of CTGF on the CXCL8/IL-8 promoter. The results showed that the activator protein-1 and nuclear factor κB binding sites of the CXCL8/IL-8 promoter are essential for CTGF-induced CXCL8/IL-8 transcription. Moreover, the CTGF-induced activation of p38 mitogen-activated protein kinase (MAPK), c-Jun-N-terminal kinase, and extracellular signal-regulated kinase (ERK) is involved in this process. In addition, adenosine-uridine-rich elements (AREs) of the CXCL8/IL-8 3'-untranslated region (3'-UTR) reduce CXCL8/IL-8 mRNA stability. To investigate whether CTGF regulates CXCL8/IL-8 gene expression at the posttranscriptional level, we transfected 293 cells with serial luciferase constructs containing different segments of the CXCL8/IL-8 3'-UTR and then stimulated the cells with CTGF. The results suggested that CTGF stabilized luciferase mRNA and increased luciferase activity by regulating the CXCL8/IL-8 3'-UTR. Moreover, the p38 MAPK pathway may contribute to CTGF-induced CXCL8/IL-8 mRNA stabilization.


Subject(s)
Connective Tissue Growth Factor/metabolism , Gene Expression Regulation , Interleukin-8/genetics , 3' Untranslated Regions , Cell Line , Connective Tissue Growth Factor/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Humans , Interleukin-8/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Monocytes/immunology , Monocytes/metabolism , NF-kappa B/metabolism , RNA Processing, Post-Transcriptional , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases/metabolism
7.
PLoS One ; 10(8): e0137177, 2015.
Article in English | MEDLINE | ID: mdl-26317424

ABSTRACT

The lymphatic endothelium plays an important role in the maintenance of tissue fluid homeostasis. It also participates in the pathogenesis of several inflammatory diseases. However, little is known about the underlying mechanisms by which lymphatic endothelial cell responds to inflammatory stimuli. In this study, we explored the mechanisms by which lipopolysaccharide (LPS) induces cyclooxygenase (COX)-2 expression in murine lymphatic endothelial cells (SV-LECs). LPS caused increases in cox-2 mRNA and protein levels, as well as in COX-2 promoter luciferase activity in SV-LECs. These actions were associated with protein phosphatase 2A (PP2A), apoptosis signal-regulating kinase 1 (ASK1), JNK1/2 and p38MAPK activation, and NF-κB subunit p65 and C/EBPß phosphorylation. PP2A-ASK1 signaling blockade reduced LPS-induced JNK1/2, p38MAPK, p65 and C/EBPß phosphorylation. Transfection with PP2A siRNA reduced LPS's effects on p65 and C/EBPß binding to the COX-2 promoter region. Transfected with the NF-κB or C/EBPß site deletion of COX-2 reporter construct also abrogated LPS's enhancing effect on COX-2 promoter luciferase activity in SV-LECs. Taken together, the induction of COX-2 in SV-LECs exposed to LPS may involve PP2A-ASK1-JNK and/or p38MAPK-NF-κB and/or C/EBPß cascade.


Subject(s)
Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Lipopolysaccharides/pharmacology , Protein Phosphatase 2/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line , Enzyme Activation/drug effects , MAP Kinase Kinase Kinase 5/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factor RelA/metabolism
8.
Int Immunopharmacol ; 28(1): 220-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26096887

ABSTRACT

Human lymphotoxin-ß receptor (LTßR), a member of the tumor necrosis factor receptor superfamily, plays an essential role in secondary lymphoid organ development, host defense, chemokine secretion, and apoptosis. In our study, LTßR activations by different stimulations were all found to induce RANTES secretion. Overexpression of LTßR or stimulation LTßR by ligands or agonistic antibody in human lung epithelial cells induced RANTES secretion However, the regulatory mechanism and the signaling cascade have not been fully elucidated. Therefore, the aim of this study was to elucidate the mechanism underlying LTßR-mediated RANTES production. Our study indicated that activation of JNK and ERK was important for the regulation of RANTES secretion. In addition, dominant negative mutants of ASK1, TAK1, and MEKK1 inhibited LTßR-induced RANTES expression. The dominant negative mutants of TRAF2, 3, and 5 also inhibited LTßR-mediated RANTES secretion. Chromatin immunoprecipitation analysis showed that LTßR activation induced the binding of c-Jun and NF-κB to the RANTES promoter. The results of this study show that LTßR activates ASK1, TAK1, and MEKK1 cascades via TRAF2, 3, and 5, resulting in the activation of JNK and ERK, which promotes the binding of c-Jun and NF-κB to the RANTES promoter, thereby increasing RANTES expression and secretion.


Subject(s)
Chemokine CCL5/biosynthesis , Lymphotoxin beta Receptor/agonists , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Janus Kinases/drug effects , Lymphotoxin beta Receptor/genetics , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/drug effects , NF-kappa B/antagonists & inhibitors , Promoter Regions, Genetic/genetics , Signal Transduction/drug effects , TNF Receptor-Associated Factor 2/genetics , Up-Regulation
9.
Int J Nanomedicine ; 9: 3631-43, 2014.
Article in English | MEDLINE | ID: mdl-25120361

ABSTRACT

Although the health effects of zinc oxide nanoparticles (ZnONPs) on the respiratory system have been reported, the fate, potential toxicity, and mechanisms in biological cells of these particles, as related to particle size and surface characteristics, have not been well elucidated. To determine the physicochemical properties of ZnONPs that govern cytotoxicity, we investigated the effects of size, electronic properties, zinc concentration, and pH on cell viability using human alveolar-basal epithelial A549 cells as a model. We observed that a 2-hour or longer exposure to ZnONPs induced changes in cell viability. The alteration in cell viability was associated with the zeta potentials and pH values of the ZnONPs. Proteomic profiling of A549 exposed to ZnONPs for 2 and 4 hours was used to determine the biological mechanisms of ZnONP toxicity. p53-pathway activation was the core mechanism regulating cell viability in response to particle size. Activation of the Wnt and TGFß signaling pathways was also important in the cellular response to ZnONPs of different sizes. The cadherin and Wnt signaling pathways were important cellular mechanisms triggered by surface differences. These results suggested that the size and surface characteristics of ZnONPs might play an important role in their observed cytotoxicity. This approach facilitates the design of more comprehensive systems for the evaluation of nanoparticles.


Subject(s)
Aluminum Oxide , Cell Survival/drug effects , Metal Nanoparticles , Proteome/drug effects , Zinc Oxide , Aluminum Oxide/chemistry , Aluminum Oxide/toxicity , Cell Line , Humans , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Proteins/analysis , Proteins/chemistry , Proteins/classification , Proteome/analysis , Proteome/chemistry , Zinc Oxide/chemistry , Zinc Oxide/toxicity
10.
Tumour Biol ; 35(7): 7147-53, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24763827

ABSTRACT

Several previous studies have investigated the association between the SULT1A1 Arg213His polymorphism and the risk of bladder cancer in various populations. However, these results remain inconsistent. Therefore, we performed this meta-analysis to evaluate the relationship between the SULT1A1 Arg213His polymorphism and the risk of bladder cancer. An extensive literature search was performed to identify all eligible studies regarding this association. The odds ratios (ORs) with 95 % confidence intervals (CIs) were used to estimate the strength of risk under fixed and random effects models. We identified and included eight case-control studies including 2,036 cases and 2,273 controls. No significant association was found between the SULT1A1 Arg213His polymorphism and the risk of bladder cancer under the dominant model; however, those with the SULT1A1 Arg/Arg genotype had a significantly increased risk (OR = 1.218, 95 % CI = 1.067-1.392, P = 0.0044) under the recessive model. In the subgroup analysis of ethnicity, a significant association was observed in Caucasians under the recessive model (OR = 1.269, 95 % CI = 1.069-1.506, P = 0.007). Furthermore, an increased risk of bladder cancer was observed between the Arg213His polymorphism and never smokers in the recessive model (OR = 1.428, 95 % CI = 1.079-1.890, P = 0.013). The results of this meta-analysis indicate that the SULT1A1 Arg213His polymorphism is associated with the risk bladder cancer under a recessive model; however, a possibly higher risk for Caucasians with the Arg/Arg genotype and never smokers needs further investigation.


Subject(s)
Arylsulfotransferase/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Urinary Bladder Neoplasms/genetics , Genotype , Humans , Polymorphism, Single Nucleotide , Risk Factors , Smoking/adverse effects , Urinary Bladder Neoplasms/pathology
11.
J Formos Med Assoc ; 112(9): 554-60, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23871550

ABSTRACT

BACKGROUND/PURPOSE: Cigarette smoking, exposure to secondhand smoke, and arsenic exposure are well known risk factors for developing urothelial carcinoma (UC). We investigated the combined effects of cigarette smoking, exposure to secondhand smoke, and the presence of urinary total arsenic on the risk of developing UC. METHODS: We conducted a hospital-based, case-control study involving 261 UC patients and 672 cancer-free control individuals between September 2002 and May 2009. RESULTS: Individuals who had smoked <100 cigarettes in their lifetime (never smokers) and had a high urinary total arsenic level (≥15.40 µg/g creatinine), and those who had smoked >100 cigarettes in their lifetime (ever smokers) and had a high urinary total arsenic level, had increased risks of developing UC (3.20-fold and 6.45-fold greater), respectively, compared to individuals who were never smokers and had a low urinary total arsenic level. Individuals who had high urinary total arsenic levels and had been exposed to secondhand smoke, and individuals with high urinary arsenic levels who had not been exposed to secondhand smoke, had increased chances (2.71-fold and 5.00-fold greater, respectively) of developing UC, compared to individuals who were not exposed to secondhand smoke and had low urinary total arsenic levels. Ever smokers who had been exposed to secondhand smoke and had a high urinary total arsenic level had the greatest increased risk for developing UC (10.82-fold greater). CONCLUSION: Individuals in a Taiwanese population who smoked cigarettes, were exposed to secondhand smoke, and a high urinary total arsenic level, had a significant risk for developing UC.


Subject(s)
Arsenic/adverse effects , Tobacco Smoke Pollution/adverse effects , Urologic Neoplasms/etiology , Adult , Aged , Arsenic/urine , Case-Control Studies , Female , Humans , Male , Methylation , Middle Aged
12.
Artif Organs ; 37(2): 211-6, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23020831

ABSTRACT

Plasmapheresis not only removes circulating antibodies but also modulates cellular immunity, including lymphocyte subsets. To investigate the effect of double-filtration plasmapheresis (DFPP) on the ratio of lymphocyte subsets in patients with myasthenia gravis (MG), we examined the percentages of B-cells, T-cells, T helper (Th) cells, T suppressor (Ts) cells, natural killer (NK) cells, NKT cells, and Th/Ts ratio before and after a single DFPP session and after a course of DFPP. A total of 26 patients were recruited; their peripheral blood lymphocyte subsets were assayed using flow cytometry. After a single session of DFPP treatment, the percentages of T-cells (P = 0.0200), Th cells (P = 0.0178), and the Th/Ts ratio (P = 0.0309) decreased significantly, whereas the percentage of NK cells (P = 0.0007) increased significantly. More importantly, after one course of DFPP treatment, the reduced clinical quantitative MG (QMG) score was correlated with the decrease of the percentage of T-cells (r = 0.5005, P = 0.0092). Fourteen thymectomized MG patients had decreased percentages of T-cells (P = 0.0304) and Th cells (P = 0.0444), whereas they had increased NK cells (P = 0.0197) after a single DFPP session. Here, transiently decreased percentages of T-cells after the full DFPP course could enhance the effectiveness of plasmapheresis for MG patients.


Subject(s)
CD3 Complex/blood , Myasthenia Gravis/therapy , Plasmapheresis , T-Lymphocyte Subsets/immunology , Adolescent , Adult , Aged , B-Lymphocytes/immunology , Biomarkers/blood , Down-Regulation , Female , Flow Cytometry , Humans , Immunosuppressive Agents/therapeutic use , Killer Cells, Natural/immunology , Lymphocyte Count , Male , Middle Aged , Myasthenia Gravis/blood , Myasthenia Gravis/immunology , Thymectomy , Time Factors , Treatment Outcome , Young Adult
13.
J Biomed Sci ; 19: 86, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-23035900

ABSTRACT

BACKGROUND: Zerumbone, a sesquiterpene compound isolated from subtropical ginger, Zingiber zerumbet Smith, has been documented to exert antitumoral and anti- inflammatory activities. In this study, we demonstrate that zerumbone induces apoptosis in human glioblastoma multiforme (GBM8401) cells and investigate the apoptotic mechanism. METHODS: We added a caspase inhibitor and transfected wild-type (WT) IKK and Akt into GBM 8401 cells, and measured cell viability and apoptosis by MTT assay and flow cytometry. By western blotting, we evaluated activation of caspase-3, dephosphorylation of IKK, Akt, FOXO1 with time, and change of IKK, Akt, and FOXO1 phosphorylation after transfection of WT IKK and Akt. RESULTS: Zerumbone (10~50 µM) induced death of GBM8401 cells in a dose-dependent manner. Flow cytometry studies showed that zerumbone increased the percentage of apoptotic GBM cells. Zerumbone also caused caspase-3 activation and poly (ADP-ribose) polymerase (PARP) production. N-benzyloxycarbonyl -Val-Ala-Asp- fluoromethylketone (zVAD-fmk), a broad-spectrum caspase inhibitor, hindered zerumbone-induced cell death. Transfection of GBM 8401 cells with WT IKKα inhibited zerumbone-induced apoptosis, and zerumbone significantly decreased IKKα phosphorylation levels in a time-dependent manner. Similarly, transfection of GBM8401 cells with Akt suppressed zerumbone-induced apoptosis, and zerumbone also diminished Akt phosphorylation levels remarkably and time-dependently. Moreover, transfection of GBM8401 cells with WT IKKα reduced the zerumbone-induced decrease in Akt and FOXO1 phosphorylation. However, transfection with WT Akt decreased FOXO1, but not IKKα, phosphorylation. CONCLUSION: The results suggest that inactivation of IKKα, followed by Akt and FOXO1 phosphorylation and caspase-3 activation, contributes to zerumbone-induced GBM cell apoptosis.


Subject(s)
Apoptosis , Forkhead Transcription Factors , I-kappa B Kinase , Oncogene Protein v-akt , Sesquiterpenes , Apoptosis/drug effects , Apoptosis/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Zingiber officinale/chemistry , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Phosphorylation , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Signal Transduction
14.
J Med Food ; 15(6): 520-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22468646

ABSTRACT

Soybean fermentation broth (SFB) exhibits potent antibacterial activity against different species of bacteria in in vitro assays and animal models. Four isoflavone compounds-daidzin, genistin, genistein, and daidzein-of SFB were analyzed and quantified by high-performance liquid chromatography. In the in vitro test, daidzin and daidzein had more potent antibacterial activity than genistin. The minimum inhibition concentration values for these bacteria of SFB ranged from 1.25% to 5%, and the minimum bactericidal concentration values of strains ranged from 2.5% to 10%, depending on the species or strain. Vancomycin-resistant Entercoccus faecalis (VRE) strains were also tested for susceptibility to SFB in two species of animal model: the Sprague-Dawley rat and the BALB/c mouse. SFB-fed Sprague-Dawley rats showed excellent elimination efficiency against VRE, close to 99% compared with the phosphate-buffered saline-fed control group. In the BALB/c mouse model, SFB antibacterial activity was 65-80% against VRE compared with the control. In conclusion, SFB contains natural antibacterial substances such as daidzin, genistin, and daidzein that inhibit bacterial growth.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Enterococcus faecalis/drug effects , Glycine max/chemistry , Gram-Positive Bacterial Infections/drug therapy , Isoflavones/therapeutic use , Soy Milk/pharmacology , Vancomycin Resistance/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Fermentation , Gram-Positive Bacterial Infections/microbiology , Isoflavones/analysis , Isoflavones/pharmacology , Male , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Rats , Rats, Sprague-Dawley
15.
J Leukoc Biol ; 87(6): 1069-82, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20200402

ABSTRACT

In this study, we investigated the role of ASK1 in PGN-induced C/EBPbeta activation and COX-2 expression in RAW 264.7 macrophages. The PGN-induced COX-2 expression was attenuated by the DNs of ASK1, JNK1, JNK2, a JNK inhibitor (SP600125), and an AP-1 inhibitor (curcumin). PGN caused ASK1 dephosphorylation time-dependently at Ser967, dissociation from the ASK1-14-3-3 complex, and subsequent ASK1 activation. In addition, PGN activated PP2A and suppression of PP2A by okadaic acid markedly inhibited PGN-induced ASK1 Ser967 dephosphorylation and COX-2 expression. PGN induced the activation of the JNK-AP-1 signaling cascade downstream of ASK1. PGN-increased C/EBPbeta expression and DNA-binding activity were inhibited by the ASK1-JNK-AP-1 signaling blockade. COX-2 promoter luciferase activity induced by PGN was attenuated in cells transfected with the COX-2 reporter construct possessing the C/EBP-binding site mutation. In addition, the ASK1-JNK-AP-1-C/EBPbeta cascade was activated in human peripheral mononuclear cells exposure to PGN. The TLR2 agonist Pam(3)CSK(4) was also shown to induce ASK1 Ser967 dephosphorylation, JNK and c-jun phosphorylation, C/EBPbeta activation, and COX-2 expression in RAW 264.7 macrophages. PGN-induced COX-2 promoter luciferase activity was prevented by selective inhibition of TLR2 and c-Jun in RAW 264.7 macrophages. Our data demonstrate that PGN might activate the TLR2-mediated PP2A-ASK1-JNK-AP-1-C/EBPbeta cascade and subsequent COX-2 expression in RAW 264.7 macrophages.


Subject(s)
Apoptosis/drug effects , Cyclooxygenase 2/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Macrophages/drug effects , Peptidoglycan/pharmacology , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Chromatin Immunoprecipitation , Humans , Immunoblotting , Immunoprecipitation , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/metabolism , Mice , Phosphorylation/drug effects , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/metabolism , Transcription Factor AP-1/metabolism
16.
J Immunol ; 182(12): 7916-27, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494316

ABSTRACT

Thrombin plays an important role in lung inflammatory diseases. Thrombin can induce connective tissue growth factor (CTGF) expression in lung fibroblasts. However, little is known about the signaling pathway in thrombin-induced CTGF expression. In this study, we investigated the role of apoptosis signal-regulating kinase 1 (ASK1) in thrombin-induced CTGF expression in human lung fibroblasts. Thrombin caused a concentration- and time-dependent increase in CTGF expression in WI-38 cells and primary lung fibroblasts. Thrombin-induced CTGF expression and CTGF-luciferase activity were inhibited by a protease-activated receptor 1 antagonist (SCH79797), the dominant-negative mutants (DNs) of ASK1 and JNK1/2, and an AP-1 inhibitor (curcumin). Thrombin caused ASK1 Ser(967) dephosphorylation, the dissociation of ASK1 and 14-3-3, and a subsequent increase in ASK1 activity. Thrombin induced increases in JNK phosphorylation and kinase activity, which were attenuated by ASK1DN. Furthermore, SCH79797 diminished the thrombin-induced ASK1 and JNK activities. Thrombin-induced CTGF-luciferase activity was predominately controlled by the sequence -747 to -184 bp upstream of the transcription start site of the human CTGF promoter and was attenuated by transfection with the deleted AP-1 binding site construct. Thrombin caused increases in c-Jun phosphorylation, the formation of an AP-1-specific DNA-protein complex, and the recruitment of c-Jun to the CTGF promoter. Furthermore, thrombin-mediated AP-1 activation was inhibited by ASK1DN, JNK1/2DN, and SP600125. These results suggest for the first time that thrombin, acting through protease-activated receptor 1, activates the ASK1/JNK signaling pathway, which in turn initiates c-Jun/AP-1 activation and recruitment of c-Jun to the CTGF promoter and ultimately induces CTGF expression in human lung fibroblasts.


Subject(s)
Connective Tissue Growth Factor/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lung/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism , Cells, Cultured , Connective Tissue Growth Factor/genetics , Enzyme Activation , Fibroblasts , Humans , Promoter Regions, Genetic/genetics , Receptors, Proteinase-Activated/metabolism , Transcription, Genetic/genetics
17.
J Biomed Sci ; 16: 43, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19405983

ABSTRACT

In the present study, we explore the role of apoptosis signal-regulating kinase 1 (ASK1) in denbinobin-induced apoptosis in human lung adenocarcinoma (A549) cells. Denbinobin-induced cell apoptosis was attenuated by an ASK1 dominant-negative mutant (ASK1DN), two antioxidants (N-acetyl-L-cysteine (NAC) and glutathione (GSH)), a c-Jun N-terminal kinase (JNK) inhibitor (SP600125), and an activator protein-1 (AP-1) inhibitor (curcumin). Treatment of A549 cells with denbinobin caused increases in ASK1 activity and reactive oxygen species (ROS) production, and these effects were inhibited by NAC and GSH. Stimulation of A549 cells with denbinobin caused JNK activation; this effect was markedly inhibited by NAC, GSH, and ASK1DN. Denbinobin induced c-Jun phosphorylation, the formation of an AP-1-specific DNA-protein complex, and Bim expression. Bim knockdown using a bim short interfering RNA strategy also reduced denbinobin-induced A549 cell apoptosis. The denbinobin-mediated increases in c-Jun phosphorylation and Bim expression were inhibited by NAC, GSH, SP600125, ASK1DN, JNK1DN, and JNK2DN. These results suggest that denbinobin might activate ASK1 through ROS production to cause JNK/AP-1 activation, which in turn induces Bim expression, and ultimately results in A549 cell apoptosis.


Subject(s)
Adenocarcinoma/enzymology , Anthraquinones/toxicity , Antineoplastic Agents/toxicity , Apoptosis , Lung Neoplasms/enzymology , MAP Kinase Kinase Kinase 5/metabolism , Phenanthrenes/toxicity , Adenocarcinoma/metabolism , Cell Death , Cell Line, Tumor , Humans , Lung Neoplasms/metabolism , Reactive Oxygen Species/metabolism
18.
J Neurosci ; 27(21): 5719-29, 2007 May 23.
Article in English | MEDLINE | ID: mdl-17522316

ABSTRACT

A pathological hallmark of Alzheimer's disease is accumulation of amyloid-beta peptide (Abeta) in senile plaques. Abeta has also been implicated in vascular degeneration in cerebral amyloid angiopathy because of its cytotoxic effects on non-neuronal cells, including cerebral endothelial cells (CECs). We explore the role of apoptosis signal-regulating kinase 1 (ASK1) in Abeta-induced death in primary cultures of murine CECs. Abeta induced ASK1 dephosphorylation, which could be prevented by selective inhibition of protein phosphatase 2A (PP2A) but not PP2B. ASK1 dephosphorylation resulted in its dissociation from 14-3-3. ASK1, released from 14-3-3 inhibition, activated p38 mitogen-activated protein kinase (p38MAPK), leading to p53 phosphorylation. p53, a proapoptotic transcription factor, in turn transactivated the expression of Bax, a proapoptotic protein. Transfection with various dominant-negative mutants (DNs), including ASK1 DN and p38MAPK DN, suppressed Abeta-induced p38MAPK activation, p53 phosphorylation, and Bax upregulation and partially prevented CEC death. Bax knockdown using a bax small interfering RNA strategy also reduced Bax expression and subsequent CEC death. These results suggest that Abeta activates the ASK1-p38MAPK-p53-Bax cascade to cause CEC death in a PP2A-dependent manner.


Subject(s)
Amyloid beta-Peptides/physiology , Apoptosis , Cerebral Cortex/enzymology , Endothelial Cells/enzymology , MAP Kinase Kinase Kinase 5/physiology , Animals , Apoptosis/genetics , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Endothelial Cells/metabolism , MAP Kinase Kinase Kinase 5/genetics , MAP Kinase Signaling System/genetics , Mice , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism
19.
J Immunol ; 177(5): 3427-38, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16920985

ABSTRACT

In this study, we examined the regulation of NF-kappaB activation and IL-8/CXCL8 expression by thrombin in human lung epithelial cells (EC). Thrombin caused a concentration-dependent increase in IL-8/CXCL8 release in a human lung EC line (A549) and primary normal human bronchial EC. In A549 cells, thrombin, SFLLRN-NH2 (a protease-activated receptor 1 (PAR1) agonist peptide), and GYPGQV-NH2 (a PAR4 agonist peptide), but not TFRGAP-NH2 (a PAR3 agonist peptide), induced an increase in IL-8/CXCL8-luciferase (Luc) activity. The thrombin-induced IL-8/CXCL8 release was attenuated by D-phenylalanyl-L-prolyl-L-arginine chloromethyl ketone (a thrombin inhibitor), U73122 (a phosphoinositide-phospholipase C inhibitor), Ro-32-0432 (a protein kinsase C alpha (PKC alpha) inhibitor), an NF-kappaB inhibitor peptide, and Bay 117082 (an IkappaB phosphorylation inhibitor). Thrombin-induced increase in IL-8/CXCL8-Luc activity was inhibited by the dominant-negative mutant of c-Src and the cells transfected with the kappaB site mutation of the IL-8/CXCL8 construct. Thrombin caused time-dependent increases in phosphorylation of c-Src at tyrosine 416 and c-Src activity. Thrombin-elicited c-Src activity was inhibited by Ro-32-0432. Stimulation of cells with thrombin activated IkappaB kinase alphabeta (IKK alphabeta), IkappaB alpha phosphorylation, IkappaB alpha degradation, p50 and p65 translocation from the cytosol to the nucleus, NF-kappaB-specific DNA-protein complex formation, and kappaB-Luc activity. Pretreatment of A549 cells with Ro-32-4032 and the dominant-negative mutant of c-Src DN inhibited thrombin-induced IKK alphabeta activity, kappaB-Luc activity, and NF-kappaB-specific DNA-protein complex formation. Further studies revealed that thrombin induced PKC alpha, c-Src, and IKK alphabeta complex formation. These results show for the first time that thrombin, acting through PAR1 and PAR4, activates the phosphoinositide-phospholipase C/PKC alpha/c-Src/IKK alphabeta signaling pathway to induce NF-kappaB activation, which in turn induces IL-8/CXCL8 expression and release in human lung EC.


Subject(s)
Chemokines, CXC/metabolism , Epithelial Cells/metabolism , Interleukin-8/metabolism , Lung/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Thrombin/metabolism , Animals , Cell Line, Tumor , Chemokines, CXC/genetics , Humans , I-kappa B Kinase/metabolism , Male , Mice , Mice, Inbred BALB C , Phosphatidylinositols/metabolism , Phosphorylation , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins pp60(c-src)/genetics , Signal Transduction
20.
Cancer Res ; 66(6): 3051-61, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16540654

ABSTRACT

TNFSF14/LIGHT is a member of the tumor necrosis factor superfamily that binds to lymphotoxin-beta receptor (LTbetaR) to induce cell death via caspase-dependent and caspase-independent pathways. It has been shown that cellular inhibitor of apoptosis protein-1 inhibits cell death by binding to LTbetaR-TRAF2/TRAF3 complexes and caspases. In this study, we found that both Kaposi's sarcoma-associated herpesvirus K7 (KSHV-K7), a viral inhibitor of apoptosis protein, and the structurally related protein survivin-DeltaEx3 could inhibit LTbetaR-mediated caspase-3 activation. However, only survivin-DeltaEx3 could protect cells from LTbetaR-mediated cell death. The differential protective effects of survivin-DeltaEx3 and KSHV-K7 can be attributed to the fact that survivin-DeltaEx3, but not KSHV-K7, is able to maintain mitochondrial membrane potential and inhibit second mitochondria-derived activator of caspase/DIABLO release. Moreover, survivin-DeltaEx3 is able to inhibit production of reactive oxygen species and can translocate from nucleus to cytosol to associate with apoptosis signal-regulating kinase 1 after activation of LTbetaR. Furthermore, survivin-DeltaEx3 protects LTbetaR-mediated cell death in caspase-3-deficient MCF-7 cells. Thus, survivin-DeltaEx3 is able to regulate both caspase-dependent and caspase-independent pathways, whereas inhibition of caspase-independent pathway is both sufficient and necessary for its protective effect on LTbetaR-mediated cell death.


Subject(s)
Apoptosis/physiology , Microtubule-Associated Proteins/physiology , Mitochondrial Proteins/physiology , Neoplasm Proteins/physiology , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Viral Proteins/physiology , Antibodies, Monoclonal/pharmacology , Apoptosis Regulatory Proteins , Carcinoma, Hepatocellular , Caspase 3 , Caspase Inhibitors , Cyclin D1/antagonists & inhibitors , Cyclin D1/metabolism , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins , Interferon-gamma/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Lymphotoxin beta Receptor , MAP Kinase Kinase Kinase 5/genetics , MAP Kinase Kinase Kinase 5/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Receptors, Tumor Necrosis Factor/agonists , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor/physiology , Recombinant Proteins , Signal Transduction , Survivin , Transfection , Tumor Necrosis Factor Ligand Superfamily Member 14 , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/physiology , Viral Proteins/genetics , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...