Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 173: 116324, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38422655

ABSTRACT

Oxidative stress (OS) is recognized as a contributing factor in the development and progression of thyroid cancer. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a pivotal transcription factor involved in against OS generated by excessive reactive oxygen species (ROS). It governs the expression of a wide array of genes implicated in detoxification and antioxidant pathways. However, studies have demonstrated that the sustained activation of Nrf2 can contribute to tumor progression and drug resistance in cancers. The expression of Nrf2 was notably elevated in papillary thyroid cancer tissues compared to normal tissues, indicating that Nrf2 may play an oncogenic role in the development of papillary thyroid cancer. Nrf2 and its downstream targets are involved in the progression of thyroid cancer by impacting the prognosis and ferroptosis. Furthermore, the inhibition of Nrf2 can increase the sensitivity of target therapy in thyroid cancer. Therefore, Nrf2 appears to be a potential therapeutic target for the treatment of thyroid cancer. This review summarized current data on Nrf2 expression in thyroid cancer, discussed the function of Nrf2 in thyroid cancer, and analyzed various strategies to inhibit Nrf2.


Subject(s)
NF-E2-Related Factor 2 , Thyroid Neoplasms , Humans , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/genetics , NF-E2-Related Factor 2/metabolism , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Oxidative Stress , Antioxidants/metabolism , Reactive Oxygen Species/metabolism
2.
iScience ; 26(8): 107477, 2023 Aug 18.
Article in English | MEDLINE | ID: mdl-37599821

ABSTRACT

Smoking carcinogen nicotine-derived nitrosamine ketone (NNK) is the most potent contributor to lung adenocarcinoma (LUAD) development, but the mechanism has not been fully elucidated. Here, we reported that fatty acid translocase CD36 was significantly overexpressed in both human LUAD tissues and NNK-induced A/J mice LUAD tumors. The overexpressed CD36 was positively correlated with Src kinase activation, smoking status, metastasis, and worse overall survival of patients with smoking history. Upon NNK binding with α7 nicotinic acetylcholine receptor (α7nAChR), sarcolemmal CD36 was increased and it interacted with surface α7nAChR and cytosol Src simultaneously, which in turn activated Src and downstream pro-carcinogenic kinase ERK1/2 and Akt, and finally caused LUAD cells to form subcutaneous and pulmonary metastatic tumors. This process could be blocked by CD36 knockdown and CD36 irreversible inhibitor SSO. Furthermore, the effect of NNK was inhibited obviously in CD36-/- A/J mice. Thus, targeting CD36 may provide a breakthrough therapy of LUAD.

3.
Theranostics ; 11(5): 2123-2136, 2021.
Article in English | MEDLINE | ID: mdl-33500715

ABSTRACT

Rationale: Hyperactivation of HGF/MET signaling pathway is a critical driver in liver tumorigenesis. Cytochrome P450 1A2 (CYP1A2) was significantly down-regulated in hepatocellular carcinoma (HCC). However, little is explored about its tumor suppressive role in HCC. In this study, we examined the functional mechanisms and clinical implication of CYP1A2 in HCC. Methods: The clinical impact of CYP1A2 was evaluated in HCC patients in Hong Kong cohort. The biological functions of CYP1A2 were investigated in vitro and in vivo. A series of biochemical experiments including Western blot assay, immunohistochemistry, quantitative reverse transcription-polymerase chain reaction, and Co-immunoprecipitation assay were conducted. Results: CYP1A2 expression was prominently silenced in HCC tumor tissues and the high expression of CYP1A2 was significantly correlated with lower AFP level, less vascular invasion, and better tumor-free survival in local cohort of HCC patients. The overexpression of CYP1A2 inhibited HCC cell viability and clonogenicity, reduced cell migration and invasion abilities in vitro, and suppressed tumorigenicity in vivo, whereas CYP1A2 knockdown exhibited the opposite effects. CYP1A2 significantly hindered HGF/MET signaling and Matrix metalloproteinases (MMPs) expression in HCC cells. Mechanically, CYP1A2 decreased HGF level and diminished HIF-1α expression, both of which are recognized as key regulators of MET activation. As the transcriptional activator of MET, HIF-1α was identified as a binding partner of CYP1A2. Direct binding of CYP1A2 with HIF-1α induced ubiquitin-mediated degradation of HIF-1α, inhibiting HIF-1α-mediated transcriptions. Conclusions: In conclusion, our results have identified CYP1A2 as a novel antagonist of HGF/MET signaling, and CYP1A2 may serve as an independent new biomarker for the prognosis of HCC patients.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/pathology , Cytochrome P-450 CYP1A2/metabolism , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/metabolism , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-met/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Cytochrome P-450 CYP1A2/genetics , Hepatocyte Growth Factor/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Proto-Oncogene Proteins c-met/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Oncogene ; 40(3): 492-507, 2021 01.
Article in English | MEDLINE | ID: mdl-33184472

ABSTRACT

Sorafenib resistance has become the main obstacle in the effective treatment of advanced hepatocellular carcinoma (HCC) patients. Activation of nuclear factor kappa B (NF-κB) is a newly identified mechanism that contributes to desensitized sorafenib. Cytochrome P450 1A2 (CYP1A2) functions as a tumor suppressor in HCC and its expression is negatively associated with NF-κB in the liver. This study aimed to study whether CYP1A2 could overcome sorafenib resistance. To investigate whether CYP1A2 and NF-κB p65 played roles in sorafenib desensitization, we established sorafenib-resistant (SR) HCC cells. SR cells decreased the expression of CYP1A2 along with the upregulation of NF-κB p65. CYP1A2 overexpression attenuated SR cell proliferation, increased sorafenib sensitivity, and inhibited the NF-κB pathway, whereas CYP1A2 silence showed opposite effects. Sorafenib, in combination with omeprazole, a CYP1A2 inducer, significantly hindered the growth and invasion of SR cells in vitro as well as decreased the tumor growth in vivo. The combination treatment markedly increased CYP1A2 expression and inhibited the sorafenib-induced NF-κB signaling. In addition, the overexpression of NF-κB p65 stimulated the SR cell growth and desensitized sorafenib in SR cells, where CYP1A2 overexpression reversed the phenomenon. Lastly, the majority of HCC tissue samples displayed decreased CYP1A2 but increased NF-κB p65 protein expression. Collectively, CYP1A2 can sensitize SR cells to sorafenib via inhibiting NF-κB p65 axis. Omeprazole in combination with sorafenib exerts a synergistic effect in alleviating acquired sorafenib resistance.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cytochrome P-450 CYP1A2/metabolism , Drug Resistance, Neoplasm , Liver Neoplasms/metabolism , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Sorafenib/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cytochrome P-450 CYP1A2/genetics , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , NF-kappa B/genetics , Neoplasm Proteins/genetics
5.
Oncogenesis ; 9(10): 97, 2020 Oct 28.
Article in English | MEDLINE | ID: mdl-33116119

ABSTRACT

The status of FOXP3 and its isoforms in hepatocellular carcinoma (HCC) is unclear. We aimed to investigate the expression and function of FOXP3 and its isoforms in HCC. The study was performed on 84 HCC patients, HCC cell lines and a mouse tumor model. The levels of FOXP3 and its isoforms were determined by nested PCR, quantitative real-time PCR and immunohistochemistry (IHC) staining. The correlation between their levels and clinicopathologic characteristics was analyzed. The full length of FOXP3 (FOXP3) and exon 3-deleted FOXP3 (FOXP3Δ3) were found to be the major isoforms in HCC. The levels of FOXP3Δ3 mRNA and protein in HCC tumor samples were not significantly different from their adjacent normal tissues. The high expression of FOXP3 protein in HCC patients showed a good overall survival. The overexpression of FOXP3 significantly reduced tumor cell proliferation, migration and invasion. The immunofluorescence result indicated that FOXP3 needed to be translocated into the nucleus to exert its inhibitory function. The luciferase assay demonstrated that FOXP3 could be synergistic with Smad2/3/4 to inhibit the oncogene c-Myc. The co-immunoprecipitation results further revealed that FOXP3 could interact with Smad2/3/4. The chromatin immunoprecipitation (ChIP) assay showed that both FOXP3 and Smad2/3/4 bound the promoter of the c-Myc to inhibit it. The in vivo mouse tumor model study confirmed the inhibitory effect of FOXP3. Collectively, the expression of tumor FOXP3 can inhibit the growth of HCC via suppressing c-Myc directly or indirectly via interacting with Smad2/3/4. Therefore, FOXP3 is a tumor suppressor in HCC.

6.
Cancer Lett ; 472: 70-80, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31874246

ABSTRACT

Liver cancer stem cells (LCSCs) initiate hepatocellular carcinoma (HCC) and contribute to its recurrence and treatment resistance. Studies have suggested ZBP-89 as a candidate tumor suppressor in HCC. We explored the role of ZBP-89 in the regulation of LCSCs. This study was performed in liver tissue samples from 104 HCC patients, 2 cell lines and mouse tumor models. We demonstrated that ZBP-89 was weakly expressed in LCSCs. Patients with high expression of LCSC markers displayed reduced survivals and higher recurrence rates after curative surgical operation. The expression of ZBP-89 was predictive for decreased recurrence. LCSC markers were negatively correlated with ZBP-89 in HCC tissues and in enriched liver tumor spheres. The exogenous expression of ZBP-89 attenuated the tumor-sphere formation and secondary colony formation capabilities of LCSCs in vitro and tumorigenicity in vivo. Furthermore, the negative effect of ZBP-89 on cancer stemness was Notch1-dependent. Localized with Notch1 intracellular domain (NICD1) in the nucleus, ZBP-89 repressed the Notch1 signaling pathway by competitive binding to NICD1 with MAML1. Collectively, ZBP-89 negatively regulates HCC stemness via inhibiting the Notch1 signaling.


Subject(s)
DNA-Binding Proteins/genetics , Liver Neoplasms/genetics , Receptor, Notch1/genetics , Transcription Factors/genetics , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Self Renewal , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/pathology , Male , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction/genetics
7.
Biochim Biophys Acta Rev Cancer ; 1871(1): 170-178, 2019 01.
Article in English | MEDLINE | ID: mdl-30630091

ABSTRACT

FOXP3 is a transcription factor, which belongs to the family of FOX protein. FOXP3 was initially discovered in regulatory T cells and supposed to play a significant role in the process of regulatory T cell differentiation. Increasing evidence has shown that FOXP3 is also expressed in tumor cells. However, the results of tumor FOXP3 is inconsistent and even the opposite. In some types of human cancers, the expression of FOXP3 is upregulated, and it can promote the development of cancers, leading to a poor prognosis. While in some other types of cancers, it is a different story. The reason for the contradictory data is unknown. The discovery of FOXP3 isoforms, interaction between tumor cells and lymphocytes in the tumor microenvironment, subcellular location, and mutation of FOXP3 may provide some clues. In this review, we first summarize and analyze the recent development. The final section focuses on the regulation of FOXP3 expression.


Subject(s)
Forkhead Transcription Factors/metabolism , Neoplasms/metabolism , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Protein Isoforms , T-Lymphocytes, Regulatory/metabolism , Tumor Microenvironment/physiology
8.
Epigenomics ; 10(4): 433-443, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29402138

ABSTRACT

AIM: Our study investigated the significance of the crosstalk between long noncoding RNAs (lncRNAs) and mRNAs in gastric cancer (GC). METHODS: lncRNA and mRNA expression profiling data in 671 GC tumors and 77 nontumorous gastric tissues were retrieved from the gene expression omnibus database: GSE54129, GSE13911, GSE19826, GSE79973, GSE15459 and GSE66229. Differentially expressed analysis, RNA coexpression network construction, gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) enrichment analyses were conducted in this study. RESULTS: Using differentially expressed and prognostic lncRNAs or mRNAs in GC, we constructed the lncRNA-mRNA coexpression networks. This network involved with vital GO and KEGG pathways. CONCLUSION: Our study reveals coexpressed lncRNAs and mRNAs as transcriptional hallmarks in GC patients which provide interesting information regarding the incidence and outcome of GC.


Subject(s)
RNA, Long Noncoding/metabolism , RNA, Messenger/metabolism , Stomach Neoplasms/genetics , Gene Expression Profiling , Kaplan-Meier Estimate , Prognosis , Stomach Neoplasms/mortality , Transcription, Genetic
9.
Epigenomics ; 9(5): 647-657, 2017 05.
Article in English | MEDLINE | ID: mdl-28517980

ABSTRACT

AIM: We performed a study to identify the role of microRNA in thymoma. PATIENTS & METHODS: One hundred twenty-three thymoma patients with clinical information and miRNA expression data from The Cancer Genome Atlas were included in the study. Comprehensive bioinformatics analysis was integrated in our analysis. RESULTS & CONCLUSION: Seven miRNAs were found to be associated with overall survival (p < 0.001). Another four miRNAs were found to be associated with disease-free survival (p < 0.001). Type C thymoma can be distinguished from nontype C thymoma by miRNAs. Interestingly, seven miRNAs showed both prognostic and subtype-specific potential. Our findings suggest that miRNAs can be used for prognostic prediction and subtype stratification.


Subject(s)
Biomarkers, Tumor/genetics , MicroRNAs/genetics , Thymoma/genetics , Thymus Neoplasms/genetics , Disease-Free Survival , Female , Humans , Male , Middle Aged , Thymoma/pathology , Thymus Neoplasms/pathology
10.
J Hepatol ; 67(2): 310-320, 2017 08.
Article in English | MEDLINE | ID: mdl-28347804

ABSTRACT

BACKGROUND & AIMS: O-GlcNAc transferase (OGT) is a unique glycosyltransferase involved in metabolic reprogramming. We investigated the functional role of OGT in non-alcoholic fatty liver disease-associated hepatocellular carcinoma (NAFLD-HCC). METHODS: The biological function of OGT in NAFLD-HCC was determined by gain- or loss- of OGT functional assays in vitro and in nude mice. OGT target factors and pathways were identified by liquid chromatography-tandem mass spectrometry (LC-MS), promoter luciferase assay, DNA binding activity assay and Western blot. RESULTS: OGT was upregulated in 12 out of 18 (66.7%) NAFLD-HCC tumor tissues by transcriptome sequencing, which was confirmed in additional NAFLD-HCC tumor tissues and cell lines. Biofunctional investigation demonstrated that OGT significantly increased cell growth (p<0.001), clonogenicity (p<0.01), migration and invasion (p<0.05) ability in vitro, and promoted xenograft tumor growth as well as lung metastasis in nude mice. The oncogenic effect of OGT was investigated, we found that OGT significantly induced palmitic acid production identified by LC-MS, which enhanced the protein expression of endoplasmic reticulum (ER) stress masters of glucose-regulated protein 78 and inositol-requiring enzyme 1α. Consequently, OGT significantly activated JNK/c-jun/AP-1 cascade by increasing protein expression of p-JNK, p-c-Jun and activation of AP-1; and induced NF-κB pathway through enhancing the protein levels of p-IKKα/ p-IKKß, p-p65, p-p50 and the NF-κB DNA binding activity. Notably, OGT inhibition by its antagonist (ST045849) suppressed cell proliferation in vitro (p<0.001) and in xenograft mice models (p<0.05). CONCLUSIONS: OGT plays an oncogenic role in NAFLD-associated HCC through regulating palmitic acid and inducing ER stress, consequently activating oncogenic JNK/c-jun/AP-1 and NF-κB cascades. LAY SUMMARY: OGT, a unique glycosyltransferase enzyme, was identified to be upregulated in non-alcoholic fatty liver disease-associated hepatocellular carcinoma tissues by transcriptome sequencing. Here, we found that OGT plays a role in cancer by promoting tumor growth and metastasis in both cell models and animal models. This effect is mediated by the induction of palmitic acid.


Subject(s)
Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , N-Acetylglucosaminyltransferases/metabolism , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation , Endoplasmic Reticulum Stress , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Biological , N-Acetylglucosaminyltransferases/genetics , Palmitic Acid/metabolism , Signal Transduction , Transplantation, Heterologous , Up-Regulation
11.
Cell Physiol Biochem ; 41(4): 1383-1392, 2017.
Article in English | MEDLINE | ID: mdl-28315871

ABSTRACT

BACKGROUND: Currently, there is a global attempt to identify potential anti-cancer agents with low toxicity. Previous studies have found that glycyrrhizin exerts anti-cancer action with low toxicity through suppressing thromboxane A2 (TxA2) in lung cancer cell lines. However, these effects have not yet been determined in animal models of lung cancer. METHODS: Human lung adenocarcinoma xenografts were established in nude mice by the introduction of A549 cells with stable transfection of the TxA2 receptor (TPα). The animal model was confirmed by the hematoxylin and eosin (H&E) method. Tumor-bearing mice were then administered graded concentrations of glycyrrhizin, cisplatin or both. After the treatments, body weights of all animals were recorded, and immunohistochemistry staining of lung tissues and serum biochemistry detection of aspartate amino transferase (AST), alanine amino transferase (ALT), urea and creatinine were carried out. RESULTS: Treatment with glycyrrhizin alone or the combination of cisplatin and glycyrrhizin profoundly reduced expression of thromboxane synthase (TxAS) as well as proliferating cell nuclear antigen (PCNA), recovered the body weight, and rescued damage of liver and kidney in tumor-bearing mice. Although it inhibited PCNA expression, cisplatin could not significantly suppress TxAS expression. Because of a positive feedback loop between TPα and TxAS, the effects of glycyrrhizin are possibly attributable to the suppression of the TxA2 pathway. CONCLUSIONS: This study provides in vivo evidence to support glycyrrhizin as a potential candidate for developing new regimens to overcome tumor progression and the resistance and toxicity of cisplatin.


Subject(s)
Adenocarcinoma/drug therapy , Glycyrrhizic Acid/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , A549 Cells , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cisplatin/pharmacology , Female , Humans , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasms, Experimental/pathology , Xenograft Model Antitumor Assays
12.
J Cancer Res Clin Oncol ; 140(3): 375-86, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24384873

ABSTRACT

BACKGROUND: Lung cancer concerns a worldwide health problem and the efficacy of available treatments is unsatisfactory. Recently, thromboxane A2 (TXA2) synthase (TXAS) and receptor (TXA2R) have been documented to play a role in lung cancer development. Therefore, dual TXA2R modulator (i.e., the dual blocker of TXAS and TXA2R) may be more efficacious to kill lung tumor cells than single TXAS inhibitor or TXA2R antagonism. The close relationship between cyclooxygenase (COX)-2 and TXAS also raises whether or how TXA2 contributes to the oncogenic activity of COX-2. This study is therefore conducted to answer these questions. METHODS: Various inhibitors and siRNA were used to evaluate the roles of TXA2 and COX-2 in the proliferation and apoptosis of lung adenocarcinoma cells. Cell proliferation was detected using both MTS ELISA and BrdU labeling ELISA. Cell cycle distribution and apoptosis were examined by flow cytometric analysis. TXB2 level, reflecting the biosynthesis of TXA2, was detected by peroxidase-labeled TXB2 conjugates using an enzyme immunoassay kit. Western blotting was performed to evaluate many biomarkers for cell cycles, apoptosis and proliferation. The levels of COXs were screened by reverse transcriptase and real-time quantitative PCR. RESULTS: We found either single TXAS inhibitor/TXA2R antagonist or the dual TXA2 modulators offered a similar inhibition on cell proliferation. Moreover, inhibition of TXA2 arrested cells at the G2/M phase and induced apoptosis. It is further demonstrated that TXA2 was able to function as a critical mediator for tumor-promoting effects of COX-2 in lung adenocarcinoma cells. CONCLUSION: The present study has for the first shown that dual TXA2 modulators and the single blocker of TXAS or TXA2R offer a similar inhibitory role in lung adenocarcinoma cell proliferation and that the tumor-promoting effects of COX-2 can largely be relayed by TXA2. Thus, TXA2 should be regarded as a critical molecule in COX-2-mediated tumor growth and a valuable target against lung cancer.


Subject(s)
Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Lung Neoplasms/metabolism , Receptors, Thromboxane/antagonists & inhibitors , Thromboxane A2/antagonists & inhibitors , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenocarcinoma/pathology , Adenocarcinoma/physiopathology , Adenocarcinoma of Lung , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Benzofurans/pharmacology , Blotting, Western , Bridged Bicyclo Compounds, Heterocyclic , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cyclooxygenase Inhibitors/therapeutic use , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Fatty Acids, Unsaturated , Flow Cytometry , Humans , Hydrazines/pharmacology , Immunoenzyme Techniques , Lung Neoplasms/pathology , Lung Neoplasms/physiopathology , Nitrobenzenes/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, Thromboxane/agonists , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sulfonamides/pharmacology , Sulfonylurea Compounds/pharmacology , Thromboxane A2/metabolism , Thromboxane-A Synthase/antagonists & inhibitors
13.
PLoS One ; 7(11): e50118, 2012.
Article in English | MEDLINE | ID: mdl-23209654

ABSTRACT

Hepatitis viral B x protein (HBx), a hepatocarcinogen, is frequently mutated. Hypoxia influences the growth of HCC and also the sensitivity of tumor cells to treatments. We aimed to test the role of HBx and acute hypoxia in the efficacy of chemotherapy. In this study, we established 4 Chang liver cell lines with the full-length HBx (HBx), the first 50 amino acids of N-terminal HBx (HBx/50), the last 104 amino acids of C-terminal HBx (HBx/51) and empty vector (CL), respectively. MTT and TNUEL assays were used to assess cell viability and apoptosis respectively. Western blot was used to determine the expression of relevant proteins. Results showed that among 4 cell lines, doxorubicin was most effective in decreasing the viability and enhancing apoptosis in HBx/51 cells, while HBx/50 cells were most resistant to the treatment. Cells in hypoxia were more susceptible to doxorubicin than cells in normoxia. Hypoxia facilitated the Bid cleavage especially in HBx/51 cells via phosphorylating p38 MAPK. p38 MAPK inhibitor significantly reduced the tBid level and increased cell viability. In conclusion, N-terminal HBx and C-terminal HBx function differentially in their ability to regulate cell growth, with the former being promotive but the latter being inhibitory. The acute hypoxia may overcome the HBx-induced resistance and facilitate the chemotherapy.


Subject(s)
Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Liver Neoplasms/virology , Liver/metabolism , Trans-Activators/physiology , Apoptosis , Cell Culture Techniques , Cell Line, Tumor , Cell Survival , Cells, Cultured , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Situ Nick-End Labeling/methods , Mutation , Protein Structure, Tertiary , Trans-Activators/chemistry , Viral Regulatory and Accessory Proteins , p38 Mitogen-Activated Protein Kinases/metabolism
14.
PLoS One ; 7(7): e41293, 2012.
Article in English | MEDLINE | ID: mdl-22829937

ABSTRACT

Polo-like kinase 4 (PLK4), belonging to serine/threonine kinase family, is critical for centriole replication and cell cycle progression. PLK4 has been proposed as a tumor suppressor in hepatocellular carcinoma (HCC). However, its expression and significance in HCC have not been well studied. In the present study, we found that PLK4 was markedly downregulated in both HCC cell lines and fresh cancer tissues, using quantitative real-time-PCR and western blot. Immunohistochemistry data also revealed that decreased expression of PLK4 was present in 72.4% (178/246) of HCC tissues, compared with the corresponding adjacent nontumorous tissues. Furthermore, PLK4 expression significantly correlated with clinicopathological parameters, including clinical stage (P=0.034), serum α-fetoprotein (AFP) (P=0.019) and tumor size (P=0.032). Moreover, HCC patients with low PLK4 expression survived shorter than those with high PLK4 expression, as indicated by overall survival (P=0.002) and disease-free survival (P=0.012) assessed by the Kaplan-Meier method. In addition, multivariate analysis suggested PLK4 as an independent predictor of overall survival (HR, 0.556; 95%CI, 0.376-0.822; P=0.003) and disease-free survival (HR, 0.547; 95%CI, 0.382-0.783; P=0.001). Collectively, our study demonstrated that PLK4 was remarkably downregulated in HCC and could be served as a potential prognostic marker for patients with this deadly disease.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Blotting, Western , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Humans , In Vitro Techniques , Liver Neoplasms/genetics , Prognosis , Protein Serine-Threonine Kinases/genetics , Real-Time Polymerase Chain Reaction
15.
PLoS One ; 7(6): e39870, 2012.
Article in English | MEDLINE | ID: mdl-22761917

ABSTRACT

Liver cancer ranks in prevalence and mortality among top five cancers worldwide. Accumulating interests have been focused in developing new strategies for liver cancer treatment. We have previously showed that dihydroartemisinin (DHA) exhibited antitumor activity towards liver cancer. In this study, we demonstrated that histone deacetylase inhibitors (HDACi) significantly augmented the antineoplastic effect of DHA via increasing apoptosis in vitro and in vivo. Inhibition of ERK phosphorylation contributed to DHA-induced apoptosis, due to the fact that inhibitor of ERK phosphorylation (PD98059) increased DHA-induced apoptosis. Compared with DHA alone, the combined treatment with DHA and HDACi reduced mitochondria membrane potential, released cytochrome c into cytoplasm, increased p53 and Bak, decreased Mcl-1 and p-ERK, activated caspase 3 and PARP, and induced apoptotic cells. Furthermore, we showed that HDACi pretreatment facilitated DHA-induced apoptosis. In Hep G2-xenograft carrying nude mice, the intraperitoneal injection of DHA and SAHA resulted in significant inhibition of xenograft tumors. Results of TUNEL and H&E staining showed more apoptosis induced by combined treatment. Immunohistochemistry data revealed the activation of PARP, and the decrease of Ki-67, p-ERK and Mcl-1. Taken together, our data suggest that the combination of HDACi and DHA offers an antitumor effect on liver cancer, and this combination treatment should be considered as a promising strategy for chemotherapy.


Subject(s)
Apoptosis/drug effects , Artemisinins/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Liver Neoplasms/pathology , Animals , Cell Line, Tumor , Cytochromes c/metabolism , Drug Synergism , Enzyme Activation , Flow Cytometry , In Situ Nick-End Labeling , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation
16.
Biochem Pharmacol ; 83(9): 1278-89, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22342732

ABSTRACT

Dihydroartemisinin (DHA), a semi-synthetic derivative of artemisinin isolated from the traditional Chinese herb Artemisia annua L., has been shown to exhibit inhibitory effects on human cancer cells. However, its antitumor ability toward hepatocellular carcinoma (HCC) has not been studied. In this study, we demonstrated that DHA significantly inhibited HCC cell growth in vitro and in vivo via inducing G2/M cell cycle arrest and apoptosis. The induction of p21 and the inhibition of cyclin B and CDC25C contributed to DHA-induced G2/M arrest. DHA-induced apoptosis was associated with mitochondrial membrane depolarization, release of cytochrome c, activation of caspases, and DNA fragmentation. Activation of caspase 9 and caspase 3, but not caspase 8, was detected in DHA-treated cells. Attenuation of apoptosis in cells pretreated with Z-VAD-FMK suggested the involvement of caspase cascade. Furthermore, p53 facilitated apoptosis caused by DHA. Bcl-2 family proteins were also responsible for DHA-induced apoptosis. DHA exposure decreased Mcl-1 expression but increased the levels of Noxa and active Bak. Bak was released from the Mcl-1/Bak complex due to the decline of Mcl-1. Further study revealed that Mcl-1 was rapidly degraded in DHA-treated cells and that DHA-induced apoptosis was largely inhibited by overexpression of Mcl-1 or RNAi-mediated decrease of Bak and Noxa. In a HCC-xenograft mouse model, the intraperitoneal injection of DHA resulted in significant inhibition of HCC xenograft tumors. Taken together, our data, for the first time, demonstrate the potential antitumor activity of DHA in HCC.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Artemisinins/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cytochromes c/metabolism , Drug Screening Assays, Antitumor , Genes, p53 , Humans , Liver Neoplasms/pathology , Mice , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/metabolism , Xenograft Model Antitumor Assays , bcl-2 Homologous Antagonist-Killer Protein/metabolism
17.
Zhongguo Zhong Yao Za Zhi ; 34(10): 1268-71, 2009 May.
Article in Chinese | MEDLINE | ID: mdl-19673393

ABSTRACT

OBJECTIVE: To investigate the effects of PsL5F (ent-11alpha-hydroxy-15-oxo-kaur-16-en-19-oic-acid, an extract from Pteris semipinnata L) on the expression of nuclear receptor subfamily 1, group D, member 1 (Nr1d1) in highly metastatic ovarian carcinoma HO-8910PM cells, and its mechanisms. METHOD: Microarray Chip was used to examine the level of Nr1d1 mRNA expression on HO-8910PM cells treated with PsL5F. Fluorescent quantitative real-time PCR assay and Western blot were performed to verify the effects of PsL5F on Nr1d1 mRNA and protein expression. RESULT: After 24 h treatment of 100 micromol x L(-1) PsL5F, the mRNA and protein levels of Nr1d1 in HO-8910PM cells were 35.34 +/- 1.07 and 7.71 +/- 0.43 times compared to those of control group (P < 0.01, P < 0.01), respectively. CONCLUSION: PsL5F can up-regulate significantly the expression of Nr1d1 in HO-8910PM cells. Antitumor effects and its mechanisms of PsL5F in HO-8910PM cells may be involved in the up-regulation of Nr1d1 expression.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Gene Expression/drug effects , Piperidones/pharmacology , Pteris/chemistry , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Humans , Neoplasm Invasiveness , Ovarian Neoplasms/pathology , RNA, Messenger/drug effects , RNA, Messenger/metabolism
18.
Cell Cycle ; 8(6): 889-95, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19221506

ABSTRACT

The cyclin-dependent kinase inhibitor p21(WAF1/CIP1) is a critical regulator of cell cycle, and it is easily degraded by proteasome through ubiquitin-dependent and -independent pathway. The mechanism of the post-translational regulation of p21 stability remains to be further clarified. In the present study, we have identified nucleophosmin (NPM)/B23, a multifunctional protein that bound p21 and contributed to its stability. The direct interaction between p21 and NPM was confirmed by reciprocal co-immunoprecipitation and GST pull-down assay. Confocal microscopy showed that NPM partially co-localized with p21 in nucleoplasm and their co-localization increased treated with Act D which induces the nucleoplasmic translocation of NPM. We observed the half life of p21 was prolonged with overexpression of NPM or Act D treatment. Knockdown of NPM by siRNA resulted in downregulation of p21 and impaired upregulation of p21 treated with Act D. Further, we examined the effect of NPM expression on the ubiquitination of p21. Overexpression of NPM inhibited the ubiquitination of p21, and depletion of NPM remarkably improved the ubiquitination of p21. Altogether, we provide evidence for a direct binding between NPM and p21, and assign NPM as a positive post-translational regulator of p21.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Nuclear Proteins/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Dactinomycin/pharmacology , Down-Regulation/drug effects , Humans , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , Nucleophosmin , Protein Stability , Protein Synthesis Inhibitors/pharmacology , RNA, Small Interfering/metabolism , Ubiquitin/metabolism , Ubiquitination/drug effects , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...