Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Technol Cancer Res Treat ; 20: 15330338211041425, 2021.
Article in English | MEDLINE | ID: mdl-34866477

ABSTRACT

Background: Liver progenitor cells (LPCs) play significant roles in the development and progression of hepatocellular carcinoma (HCC). However, no studies on the value of LPC-related genes for evaluating HCC prognosis exist. We developed a gene signature of LPC-related genes for prognostication in HCC. Methods: To identify LPC-related genes, we analyzed mRNA expression arrays from a dataset (GSE57812 & GSE 37071) containing LPCs, mature hepatocytes, and embryonic stem cell samples. HCC RNA-Seq data from The Cancer Genome Atlas (TCGA) were used to explore the differentially expressed genes (DEGs) related to prognosis through DEG analysis and univariate Cox regression analysis. Lasso and multivariate Cox regression analyses were performed to construct the LPC-related gene prognostic model in the TCGA training dataset. This model was validated in the TCGA testing set and an external dataset (International Cancer Genome Consortium [ICGC] dataset). Finally, we investigated the relationship between this prognostic model with tumor-node-metastasis stage, tumor grade, and vascular invasion of HCC. Results: Overall, 1770 genes were identified as LPC-related genes, of which 92 genes were identified as DEGs in HCC tissues compared with normal tissues. Furthermore, we randomly assigned patients from the TCGA dataset to the training and testing cohorts. Twenty-six DEGs correlated with overall survival (OS) in the univariate Cox regression analysis. Lasso and multivariate Cox regression analyses were performed in the TCGA training set, and a 3-gene signature was constructed to stratify patients into 2 risk groups: high-risk and low-risk. Patients in the high-risk group had significantly lower OS than those in the low-risk group. Receiver operating characteristic curve analysis confirmed the signature's predictive capacity. Moreover, the risk score was confirmed to be an independent predictor for patients with HCC. Conclusion: We demonstrated that the LPC-related gene signature can be used for prognostication in HCC. Thus, targeting LPCs may serve as a therapeutic alternative for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Stem Cells , Transcriptome , AMP-Activated Protein Kinases/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/secondary , Databases, Genetic , Enhancer of Zeste Homolog 2 Protein/genetics , Female , Gene Expression Profiling , Humans , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness/genetics , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Osteopontin/genetics , Prognosis , Proportional Hazards Models , RNA, Messenger/metabolism , ROC Curve , Retrospective Studies , Risk Assessment/methods , Survival Rate , Young Adult
2.
J Cancer ; 10(25): 6252-6260, 2019.
Article in English | MEDLINE | ID: mdl-31772658

ABSTRACT

BACKGROUND: Phosphodiesterase 4D (PDE4D) has recently been reported as an oncogene in various types of human cancers. However, the expression and significance of PDE4D in pancreatic ductal adenocarcinoma (PDAC) have not been elucidated. Methods: Immunohistochemistry (IHC) was used to examine the expression of PDE4D in 104 clinicopathologically characterized PDAC cases. PDE4D expression in paired tumor tissues and adjacent noncancerous tissues were detected by western blotting and real time qRT-PCR. The correlation of PDE4D expression levels with clinicopathological features and prognosis in patients were analyzed by univariate and multivariate methods. Effect of PDE4D on pancreatic cancer cells was detected by cell migration and invasion assays. Results: We found that PDE4D was significantly up-regulated in PDAC tumor tissues compared to those paired adjacent noncancerous tissues at both protein and mRNA levels. High level of PDE4D was significantly associated with clinical stage (P = 0.004), T classification (P = 0.003), lymph node metastasis (P = 0.022) and liver metastasis (P = 0.038). Patients with higher levels of PDE4D had shorter overall survival time contrast with those with lower PDE4D expression (P = 0.002). Multivariate analysis indicated that PDE4D may be an independent prognostic factor for PDAC. PDE4D depletion significantly suppressed ß-catenin and Snail expression as well as the migration and invasion abilities of pancreatic cancer cells. Conclusions: Our study reveals that PDE4D up-regulated in PDAC was closely associated with poor prognosis of PDAC patients and multiple aggressive clinicopathological characteristics. PDE4D could be a useful prognostic biomarker and therapeutic target for PDAC.

3.
J Biomed Nanotechnol ; 15(8): 1812-1823, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31219012

ABSTRACT

Golgi phosphoprotein 3 (GOLPH3), a proto-oncogene product, is significantly increased during the progression of several types of cancer. However, its biological role and underlying mechanism in the development of renal cell carcinoma (RCC) remain poorly understood. In this study, GOLPH3 was found to be highly expressed in RCC specimens compared to the corresponding non-tumor tissues. In vitro, ectopic overexpression of GOLPH3 substantially promoted the proliferative and invasive capacity of RCC cells, while the depletion of GOLPH3 significantly inhibited proliferation and invasion of RCC cells. Furthermore, the average tumor volume was significantly increased in mice injected with 769-P cells highly expressing GOLPH3, whereas GOLPH3 knockdown reduced the tumor growth rate. Mechanistically, using a high-throughput phospho-proteome array verified by Western blotting, we have identified that phosphorylated proteins (FAK, Raf1, MEK, and GSK3ß) were upregulated, activating, in turn, FAK/Raf1/MEK and Wnt/ß-catenin signaling pathways in RCC cells. Taken together, our findings demonstrate that GOLPH3, whose expression is related to enhanced cell proliferation and invasion via activation of GOLPH3-FAK/Raf1/MEK axis or Wnt/ß-catenin signaling pathways, may provide a new therapeutic target to treat renal cell carcinoma.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Membrane Proteins/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Mice , Phenotype , Phosphoproteins , Proto-Oncogene Mas , Wnt Signaling Pathway
4.
J Cell Biochem ; 120(3): 4385-4397, 2019 03.
Article in English | MEDLINE | ID: mdl-30260040

ABSTRACT

Acetaminophen (APAP) is a widely used over-the-counter analgesic and antipyretic. It can cause hepatotoxicity. Recent studies demonstrated that hydrogen sulfide (H2 S) exhibits cell protection in several cell types. This study was designed to investigate whether H 2 S ameliorated APAP-induced acute liver injury and to elucidate its mechanisms. In this study, we analyzed the detailed biological and molecular processes of APAP-induced hepatotoxicity using a bioinformatics analysis, which showed that apoptosis and the c-Jun N-terminal kinase (JNK)/mitogen-activated protein kinase pathway were confirmed to play critical roles in these processes. We further investigated the protective effects of H 2 S on APAP-induced hepatotoxicity. In vivo, we observed that the exogenous supplement of H 2 S ameliorated APAP-induced liver injury. Cystathionine-ß-synthase (CBS) and cystathionine-γ-lyase (CSE) systems were the endogenous pathway of H 2 S. The expression of CBS/CSE was decreased in APAP-treated mice, while H 2 S could significantly restore it. In addition, APAP-induced JNK activation was inhibited by H 2 S in vivo. In vitro, H 2 S abolished the active effects of APAP on caspase3, Bax, and Bcl-2 expressions as well as JNK phosphorylation in hepatocytes. It was found through flow cytometry that the amount of APAP-induced apoptotic hepatocytes was decreased in the presence of H 2 S. In conclusion, our results suggested that H 2 S attenuated APAP-induced apoptosis in hepatocytes through JNK/MAPK siganaling pathway.


Subject(s)
Acetaminophen/adverse effects , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes/metabolism , MAP Kinase Signaling System/drug effects , Acetaminophen/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Line , Chemical and Drug Induced Liver Injury/pathology , Hepatocytes/pathology , Humans , Hydrogen Sulfide , MAP Kinase Kinase 4/metabolism , Male , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism
5.
Oncol Rep ; 40(5): 2628-2636, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30226561

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a pro­inflammatory cytokine that serves important roles in cancer. MIF overexpression is frequently observed in numerous human cancer types, including pancreatic carcinoma. However, the prognostic value and function of MIF in pancreatic ductal adenocarcinoma (PDAC) have not been fully elucidated. In the present study, upregulation of MIF expression in PDAC tissue compared with adjacent normal tissue was observed. Furthermore, MIF overexpression was identified to be significantly associated with poor survival rates in patients with PDAC. Multivariate Cox regression analysis confirmed that MIF was an independent risk factor for poor survival. Functional analyses demonstrated that MIF knockdown significantly inhibited the proliferation and invasion of pancreatic cancer cells in vitro compared with control cells. IN addition, mechanistic investigations revealed that silencing MIF leads to inhibition of AKT serine/threonine kinase and extracellular­signal­regulated kinase activation, and suppression of cyclin D1 and matrix metalloproteinase­2 expression, which may suppress tumor proliferation and invasion. These results highlight the importance of MIF overexpression in PDAC aggressiveness, and indicate that MIF may be a potential therapeutic target for pancreatic cancer.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/genetics , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Adenocarcinoma/pathology , Adult , Aged , Apoptosis/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cyclin D1/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Matrix Metalloproteinase 2/genetics , Middle Aged , Neoplasm Metastasis , Prognosis , Progression-Free Survival
6.
Dig Liver Dis ; 47(1): 62-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25277864

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptor δ (PPARδ) is a versatile regulator of distinct biological processes and overexpression of PPARδ in cancer may be partially related to its suppression of its own co-regulators. AIMS: To determine whether recruited suppressor proteins bind to and regulate PPARδ expression, activity and PPARδ-dependent cholangiocarcinoma proliferation. METHODS: Yeast two-hybrid assays were done using murine PPARδ as bait. PPARδ mRNA expression was determined by qPCR. Protein expression was measured by western blot. Immunohistochemistry and fluorescence microscopy were used to determine PPARδ expression and co-localization with NDP Kinase alpha (NM23-H2). Cell proliferation assays were performed to determine cell numbers. RESULTS: Yeast two-hybrid screening identified NM23-H2 as a PPARδ binding protein and their interaction was confirmed. Overexpressed PPARδ or treatment with the agonist GW501516 resulted in increased cell proliferation. NM23-H2 siRNA activated PPARδ luciferase promoter activity, upregulated PPARδ RNA and protein expression and increased GW501516-stimulated CCA growth. Overexpression of NM23-H2 inhibited PPARδ luciferase promoter activity, downregulated PPARδ expression and AKT phosphorylation and reduced GW501516-stimulated CCA growth. CONCLUSIONS: We report the novel association of NM23-H2 with PPARδ and the negative regulation of PPARδ expression by NM23-H2 binding to the C-terminal region of PPARδ. These findings provide evidence that the metastasis suppressor NM23-H2 is involved in the regulation of PPARδ-mediated proliferation.


Subject(s)
Bile Duct Neoplasms/genetics , Bile Ducts, Intrahepatic , Cholangiocarcinoma/genetics , Gene Expression Regulation, Neoplastic , NM23 Nucleoside Diphosphate Kinases/genetics , PPAR gamma/genetics , RNA, Messenger/metabolism , Animals , Bile Duct Neoplasms/metabolism , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Cholangiocarcinoma/metabolism , Down-Regulation , Humans , Immunohistochemistry , Mice , NM23 Nucleoside Diphosphate Kinases/metabolism , PPAR gamma/metabolism , RNA, Small Interfering , Rats , Reverse Transcriptase Polymerase Chain Reaction , Yeasts
7.
Oncotarget ; 5(14): 5570-80, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25015194

ABSTRACT

Macrophage migration inhibitory factor (MIF), a proinflammatory and immunoregulatory chemokine, plays important roles in cancer-related biological processes. However, few studies have focused on the clinical relevance of MIF and cyclin D1 expression in hepatocellular carcinoma cells (HCCs). In this study, MIF and cyclin D1 expression levels in HCC tissues and cell lines were significantly upregulated compared with adjacent normal tissues or a normal liver cell line. In HCC specimens, MIF expression positively correlated with cyclin D1 expression. Additionally, MIF and cyclin D1 expression positively correlated with tumor size. MIF knockdown inhibited the proliferation of PLC and HepG2 cells and promoted apoptosis. However, small interfering RNA (siRNA) against MIF did not influence the cell cycle in these cells. In an in vivo xenograft model, MIF knockdown reduced the tumor growth rate. The expression levels of Bcl-2, p-caspase-3, BIM and Bax were upregulated, while the expression levels of cyclin D1, p-Akt and p-ERK were downregulated in MIF-knockdown cells. These findings indicate that MIF siRNA reduces proliferation and increases apoptosis in HCC cells. MIF knockdown inhibits the expression of growth-related proteins and induces the expression of apoptosis-related proteins, supporting a role for MIF as a novel therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/therapy , Cyclin D1/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/therapy , Macrophage Migration-Inhibitory Factors/deficiency , RNA, Small Interfering/genetics , Animals , Apoptosis/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Female , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Macrophage Migration-Inhibitory Factors/genetics , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , RNA, Small Interfering/administration & dosage , Xenograft Model Antitumor Assays
8.
J Transl Med ; 12: 33, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24495516

ABSTRACT

BACKGROUND: According to cancer-related microRNA (miRNA) expression microarray research available in public databases, miR-362 expression is elevated in gastric cancer. However, the expression and biological role of miR-362 in gastric progression remain unclear. METHODS: miR-362 expression levels in gastric cancer tissues and cell lines were determined using real-time PCR. The roles of miR-362, in promoting gastric cancer cell proliferation and apoptosis resistance, were assessed by different biological assays, such as colony assay, flow cytometry and TUNEL assay. The effect of miR-362 on NF-κB activation was investigated using the luciferase reporter assay, fluorescent immunostaining. RESULTS: MiR-362 overexpression induced cell proliferation, colony formation, and resistance to cisplatin-induced apoptosis in BGC-823 and SGC-7901 gastric cancer cells. MiR-362 increased NF-κB activity and relative mRNA expression of NF-κB-regulated genes, and induced nuclear translocation of p65. Expression of the tumor suppressor CYLD was inhibited by miR-362 in gastric cancer cells; miR-362 levels were inversely correlated with CYLD expression in gastric cancer tissue. MiR-362 downregulated CYLD expression by binding its 3' untranslated region. NF-κB activation was mechanistically associated with siRNA-mediated downregulation of CYLD. MiR-362 inhibitor reversed all the effects of miR-362. CONCLUSION: The results suggest that miR-362 plays an important role in repressing the tumor suppressor CYLD and present a novel mechanism of miRNA-mediated NF-κB activation in gastric cancer.


Subject(s)
Apoptosis/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Signal Transduction/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , 3' Untranslated Regions/genetics , Base Sequence , Cell Line, Tumor , Cell Proliferation , Deubiquitinating Enzyme CYLD , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Molecular Sequence Data , Tumor Suppressor Proteins/metabolism , Up-Regulation/genetics
9.
J Biol Chem ; 289(7): 4262-72, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24375408

ABSTRACT

Transient Receptor Potential mucolipin (TRPML) channels are implicated in endolysosomal trafficking, lysosomal Ca(2+) and Fe(2+) release, lysosomal biogenesis, and autophagy. Mutations in human TRPML1 cause the lysosome storage disease, mucolipidosis type IV (MLIV). Unlike vertebrates, which express three TRPML genes, TRPML1-3, the Drosophila genome encodes a single trpml gene. Although the trpml-deficient flies exhibit cellular defects similar to those in mammalian TRPML1 mutants, the biophysical properties of Drosophila TRPML channel remained uncharacterized. Here, we show that transgenic expression of human TRPML1 in the neurons of Drosophila trpml mutants partially suppressed the pupal lethality phenotype. When expressed in HEK293 cells, Drosophila TRPML was localized in both endolysosomes and plasma membrane and was activated by phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2) applied to the cytoplasmic side in whole lysosomes and inside-out patches excised from plasma membrane. The PI(3,5)P2-evoked currents were blocked by phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), but not other phosphoinositides. Using TRPML A487P, which mimics the varitint-waddler (Va) mutant of mouse TRPML3 with constitutive whole-cell currents, we show that TRPML is biphasically regulated by extracytosolic pH, with an optimal pH about 0.6 pH unit higher than that of human TRPML1. In addition to monovalent cations, TRPML exhibits high permeability to Ca(2+), Mn(2+), and Fe(2+), but not Fe(3+). The TRPML currents were inhibited by trivalent cations Fe(3+), La(3+), and Gd(3+). These features resemble more closely to mammalian TRPML1 than TRPML2 and TRPML3, but with some obvious differences. Together, our data support the use of Drosophila for assessing functional significance of TRPML1 in cell physiology.


Subject(s)
Cell Membrane/metabolism , Drosophila Proteins/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Metals/metabolism , Phosphatidylinositol Phosphates/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Cations/metabolism , Cell Membrane/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Endosomes/genetics , HEK293 Cells , Humans , Ion Transport/physiology , Lysosomes/genetics , Mutation, Missense , Phosphatidylinositol Phosphates/genetics , Transient Receptor Potential Channels/genetics
10.
J Pathol ; 227(4): 470-80, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22450659

ABSTRACT

SPHK1 expression is elevated in gastric cancer and is associated with shorter survival times for patients. However, the molecular mechanism of SPHK1 up-regulation in gastric cancer remains unclear. In the present study, we report that miR-124 down-regulated SPHK1 expression by directly targeting its 3'-untranslated region (3'-UTR) and that miR-124 expression was inversely correlated with SPHK1 expression in gastric cancer samples. Furthermore, we demonstrated that, similar to the effect of silencing SPHK1, up-regulation of miR-124 markedly inhibited proliferation and tumourigenicity of gastric cancer cells both in vitro and in vivo. This was found to be mechanistically associated with induction of cyclin-dependent kinase inhibitors p21$^{{\rm Cip1}}$ and p27$^{{\rm Kip1}}$, enhancement of the transcriptional activity of FOXO1 and suppression of AKT activity. Moreover, we showed that the re-introduction of SPHK1 (without the 3'-UTR), but not with the 3'-UTR, could abrogate the miR-124-mediated induction of p21$^{{\rm Cip1}}$ and p27$^{{\rm Kip1}}$, as well as rescue the miR-124-induced proliferation inhibition. Together, these results suggest that miR-124 has an important role in the suppression of gastric cancer and presents a novel mechanism of miRNA-mediated SPHK1 expression in cancer cells.


Subject(s)
Adenocarcinoma/pathology , Cell Proliferation , Down-Regulation/physiology , MicroRNAs/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Stomach Neoplasms/pathology , Adenocarcinoma/physiopathology , Cell Cycle/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic , Cyclin-Dependent Kinase Inhibitor p21/physiology , Cyclin-Dependent Kinase Inhibitor p27/physiology , Forkhead Box Protein O1 , Forkhead Transcription Factors/physiology , Humans , In Vitro Techniques , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Stomach Neoplasms/physiopathology
11.
Pancreas ; 41(4): 629-35, 2012 May.
Article in English | MEDLINE | ID: mdl-22249132

ABSTRACT

OBJECTIVES: Oncogenic transcription factor forkhead box M1 (FoxM1)-related clinicopathologic characteristics and prognosis of patients with pancreatic ductal adenocarcinoma (PDA) have not been identified. Our aim of studying FoxM1 expression level and survival rate of PDA is to determine whether FoxM1 is a valuable prognostic predictor for PDA patients. METHODS: Expressional levels of FoxM1 mRNA and protein in paired pancreatic cancer lesions and adjacent noncancerous tissues were examined by reverse transcription-polymerase chain reaction and Western blotting. FoxM1 expression was analyzed by immunohistochemistry in 80 patients with PDA. The correlations between FoxM1 immunostaining levels and clinicopathologic factors, as well as the follow-up data of patients, were analyzed statistically. RESULTS: FoxM1 protein and mRNA levels were elevated in pancreatic carcinoma lesions compared with the paired adjacent noncancerous tissues. A high level of expression of FoxM1 was significantly correlated with clinical staging (P = 0.004), lymph node metastasis (P = 0.009), and histological differentiation (P = 0.017). Patients with a higher FoxM1 expression had a significantly shorter survival time than those patients with lower FoxM1 expression (P < 0.001). The multivariate analysis revealed that FoxM1 could serve as an independent factor of poor prognosis. CONCLUSIONS: Our finding indicates that FoxM1 could be used as prognostic molecular marker and therapeutic target for PDA.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Forkhead Transcription Factors/metabolism , Pancreatic Neoplasms/metabolism , Adult , Aged , Blotting, Western , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/surgery , Female , Follow-Up Studies , Forkhead Box Protein M1 , Humans , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Pancreas/metabolism , Pancreas/pathology , Pancreatectomy , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Up-Regulation
12.
Zhonghua Gan Zang Bing Za Zhi ; 17(6): 422-5, 2009 Jun.
Article in Chinese | MEDLINE | ID: mdl-19567019

ABSTRACT

OBJECTIVE: To investigate the effect of osteopontin (OPN) on the invasion and metastasis of human hapatocellular carcinoma (HCC). METHODS: HCC cell lines (HCC-LM3) were transfected with the chemically synthesized small interfering RNA (siRNA). Real-time PCR and Western blot were used to quantify the mRNA and OPN protein levels. The malignant phenotypes including cellular growth, colony formation and invasion capability of the HCC cells were analyzed. RESULTS: The OPN mRNA and proteins levels were decreased by 75% and 80% in OPN siRNA treated cells. Colony formation and migratory capability were reduced in OPN siRNA treated cells (P < 0.05). CONCLUSION: The specific siRNA is able to reduce the OPN expression at both the mRNA and protein levels and significantly inhibits the invasiveness of HCC cells.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Osteopontin/genetics , RNA, Small Interfering/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Genetic Vectors , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/prevention & control , Osteopontin/antagonists & inhibitors , Osteopontin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
13.
Zhonghua Wai Ke Za Zhi ; 47(5): 369-72, 2009 Mar 01.
Article in Chinese | MEDLINE | ID: mdl-19595017

ABSTRACT

OBJECTIVE: To investigate the stimulated effect of liver regeneration on colon cancer cells in remnant liver in rats. METHODS: Rat models with liver metastases or retro-peritoneal metastases of colon cancer were established: animals underwent 37% or 70% liver resection and were compared with a sham laparotomy (15, 25, 15 cases, respectively). Metastases were performed two weeks before resection. Rats were killed 3 weeks after the resection. Total body weight, liver and tumor weights were recorded. The human colon adenocarcinoma cell line Lovo was cultured in the presence of portal serum withdrawn 24 hours and 14 days after partial hepatectomy (PH). DNA synthesis was assessed by flow cytometry analysis for 5-Bromodeoxyuridine (5-BrdU) incorporation. RESULTS: The tumor growth was accelerated in the remnant liver in 70% PH group, but the tumors in 37% PH group and retro-peritoneal site were not influenced by PH. Compared with the control group, after cultured 72 hours with portal serum withdrawn 24 h after PH, a higher 5-BrdU incorporation was found in the Lovo cell lines (P < 0.05), and it reached the peak after 120 hours of culture (P < 0.05). No difference was found between the groups when cultured with the portal serum withdrawn 14 d after PH (P > 0.05). CONCLUSIONS: PH may accelerate the growth of residual microscopic tumor in the liver which contributes to local recurrence. It has no systemic effect and effects on the cancer cell lines in extrahepatic sites. The excision extension is related to the stimulating effects on the cancer cell line, and subtotal hepatectomy is presumably a major determinant.


Subject(s)
Colonic Neoplasms/pathology , Hepatectomy , Liver Neoplasms, Experimental/secondary , Liver/physiopathology , Animals , Cell Line, Tumor , Colonic Neoplasms/physiopathology , Humans , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/surgery , Liver Regeneration , Neoplasm Recurrence, Local , Rats , Rats, Wistar , Xenograft Model Antitumor Assays
14.
Zhonghua Gan Zang Bing Za Zhi ; 17(2): 102-6, 2009 Feb.
Article in Chinese | MEDLINE | ID: mdl-19254456

ABSTRACT

OBJECTIVES: To observe the expression of macrophage migration inhibition factor (MIF) and p27 in hepatocellular carcinoma tissue, and to investigate the effect of MIF on the expression of p27 in hepatocellular carcinoma (HCC) cells. METHODS: Immunohistochemistry and quantitative RT-PCR were performed to detect the expression of MIF and p27 in HCC tissues and peri-tumor tissues. Specific small interfering RNA (siRNA) targeting MIF gene was chemically synthesized and then transfected at the concentration of 50 nmol/L and 100 nmol/L into PLC cells and Hep3B cells. The mRNA levels of MIF and p27 after MIF siRNA treatment were quantified by real-time RT-PCR. RESULTS: MIF protein and mRNA were over-expressed in the HCC tumor tissues compared to these in the peri-tumor tissues (P less than 0.01). The expression of p27 protein and mRNA was significantly lower in the HCC tumor tissues compared to these in the peri-tumor tissues (P less than 0.01). Compared to normal liver cell line L-02, HCC cell lines expressed higher level of MIF (F=61.036, P less than 0.01) and lower level of p27 (F=529.853, P less than 0.01). In MIF siRNA treated PLC and Hep3B cells, the MIF mRNA was decreased in a dose-dependent manner (F=320.1, P less than 0.01; F=201.2, P less than 0.01). The p27 mRNA was significantly up-regulated in MIF siRNA treated PLC and Hep3B cells compared to control siRNA transfected cells (F=419.4, P less than 0.01; F=459.9, P less than 0.01). CONCLUSIONS: MIF is over-expressed in HCC tumor tissues, and the expression of p27 is repressed by MIF.


Subject(s)
Carcinoma, Hepatocellular , Macrophage Migration-Inhibitory Factors , Cell Line, Tumor , Humans , Immunohistochemistry , Liver Neoplasms , RNA, Messenger/genetics
15.
Clin Cancer Res ; 15(4): 1393-9, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19228740

ABSTRACT

PURPOSE: The present study was to investigate the clinical significance of sphingosine kinase 1 (SPHK1), an oncoenzyme, in the development and progression of gastric cancer. EXPERIMENTAL DESIGN: mRNA and protein levels of SPHK1 expression in normal gastric epithelial cells, gastric cancer cell lines, and paired gastric cancer lesions and the adjacent noncancerous tissues were examined using reverse transcription-PCR and Western blotting. Immunohistochemistry was employed to analyze SPHK1 expression in 175 clinicopathologically characterized gastric cancer cases. Statistical analyses were applied to derive prognostic and diagnostic associations. RESULTS: Levels of SPHK1 mRNA and protein were higher in gastric cancer cell lines than in normal gastric epithelial cells. SPHK1 protein level was up-regulated in gastric cancer lesions compared with that in the paired adjacent noncancerous tissues. Gastric cancer tissues from 115 of 175 (65.7%) patients revealed high level of SPHK1 protein expression in contrast to the undetectable or marginally detectable expression of SPHK1 in the adjacent noncancerous gastric tissues. Significantly different expression levels of SPHK1 were found in patients at different clinical stages (P=0.003), T classification (P=0.035), and M classification (P=0.020). Patients with higher SPHK1 expression had shorter overall survival time, whereas those with lower SPHK1 expression survived longer. Further multivariate analysis suggested that SPHK1 up-regulation was an independent prognostic indicator for the disease. CONCLUSIONS: SPHK1 protein could be a useful marker for the prognosis of gastric cancer. Further study on the potential use of SPHK1 as a therapeutic target is also warranted.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor)/analysis , Stomach Neoplasms/enzymology , Adult , Aged , Biomarkers, Tumor/analysis , Cell Line, Tumor , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Phosphotransferases (Alcohol Group Acceptor)/genetics , Prognosis , RNA, Messenger/analysis , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology
16.
Zhonghua Yi Xue Za Zhi ; 88(26): 1846-50, 2008 Jul 08.
Article in Chinese | MEDLINE | ID: mdl-19040022

ABSTRACT

OBJECTIVE: To evaluate the impact of the recombined adeno-associated virus encoding soluble tumor necrosis factor related apoptosis inducing ligand gene (rAAV-sTRAIL) on proliferation and apoptosis of human hepatocellular carcinoma (HCC) cells, and to investigate the feasibility and efficiency of transfection of rAAV-sTRAIL into human HCC cells by ultrasound microbubble intensifier. METHODS: Human HCC cells of the line HepG2 were transfected with rAAV-sTRAIL or rAAV-sTRAIL combined with microbubble echocontrast agent and appropriate dose of ultrasound irradiation. RT-PCR and Western blotting were used to detect the mRNA and protein expression of sTRAIL gene. MTT method was used to detect the proliferation inhibition rate, and the apoptosis rate of the HepG2 cells was evaluated by flow cytometry. RESULTS: The expression levels of sTRAIL mRNA and protein were higher in the rAAV-TRAIL combined with ultrasound microbubble group than in the rAAV-sTRAIL group (both P < 0.05). The proliferation inhibition rate of the rAAV-TRAIL combined with ultrasound microbubble group was significantly higher than that of the rAAV-sTRAIL group (P < 0.05). The apoptotic effect of the rAAV-TRAIL combined with ultrasound microbubble group was greater than that of the rAAV-sTRAIL group (P < 0.05). CONCLUSION: TRAIL has a potential role to inhibit e proliferation and induce apoptosis of human hepatocellular carcinoma cells. Ultrasound and microbubble echocontrast agent increase the transfection rate of rAAV vector into HCC cells.


Subject(s)
Dependovirus/genetics , TNF-Related Apoptosis-Inducing Ligand/physiology , Transfection/methods , Apoptosis/drug effects , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Contrast Media/administration & dosage , Genetic Vectors , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Microbubbles , Reverse Transcriptase Polymerase Chain Reaction , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Ultrasonics
17.
Zhonghua Zhong Liu Za Zhi ; 30(7): 511-4, 2008 Jul.
Article in Chinese | MEDLINE | ID: mdl-19062717

ABSTRACT

OBJECTIVE: To investigate the expression of macrophage migration inhibitory factor (MIF), p16 and vascular endothclial growth factor (VEGF) proteins and their relationship with clinicopathological features in cervical cancer. METHODS: Tissue microarray (TMA) and immunohistochemistry were used to detect the expression of MIF, p16 and VEGF proteins in specimens of 10 normal cervical epithelial tissues, 18 cervical intraepithelial neoplasia (CIN II, III) and 31 cervical squamous cell carcinomas. Western blotting was used to detect the expression of MIF, p16 and VEGF proteins in fresh samples of 3 normal cervical epithelial tissues, 3 CIN (III) and 6 cervical squamous cell carcinomas (3 Ib and 3 IIb). RESULTS: Positive expression rates of MIF were 0, 72.2% and 93.5% in the normal, CIN and carcinoma samples, 20.0%, 33.3% and 71.0% for p16, and 10.0%, 44.4% and 74.2% for VEGF, respectively. The expression rates and levels of the three genes were significantly higher in cervical carcinomas than those in CIN. MIF expression was significantly higher in the cases with lower differentiation (17 cases, P = 0.021), and was positively correlated with VEGF expression (P = 0.0045). VEGF expression rate was significantly higher in both cases of poorly differentiated carcinomas and those with stage II b carcinoma or beyond (P = 0.004, P = 0.008). p16 expression was not found to be correlated with tumor differentiation or clinical stage. It was showed by Western blotting that the expression levels of MIF, VEGF and p16 were significantly higher in the carcinomas than those in CIN or normal tissues. CONCLUSION: Expression of MIF, VEGF and p16 are probably involved in the process of cervical carcinogenesis. MIF expression is correlated with tumor differentiation. VEGF expression is correlated with both tumor differentiation and clinical stage.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Neoplasm Proteins/metabolism , Uterine Cervical Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism , Carcinoma, Squamous Cell/pathology , Cervix Uteri/metabolism , Cervix Uteri/pathology , Cyclin-Dependent Kinase Inhibitor p16 , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Staging , Uterine Cervical Neoplasms/pathology , Uterine Cervical Dysplasia/metabolism , Uterine Cervical Dysplasia/pathology
20.
Zhonghua Gan Zang Bing Za Zhi ; 16(3): 184-7, 2008 Mar.
Article in Chinese | MEDLINE | ID: mdl-18364076

ABSTRACT

OBJECTIVE: To study the relationship between the dynamic changes of caveolin-1 with the degrees of liver fibrosis and portal venous pressure (PVP) in the process of rat liver cirrhosis formation induced by dimethylnitrosamine (DMN); also to investigate the mechanisms of caveolin-1 in the regulation of endothelial nitric oxide synthase (eNOS). METHODS: Liver cirrhosis was induced in rats by DMN. The degrees of liver fibrosis and PVP were measured. NOS activity was assessed by citrulline generation. Protein expressions of caveolin-1, eNOS and caveolin-1-eNOS interactions were examined by Western blot and immunoprecipitation, respectively. RESULTS: Four weeks after DMN administration, liver fibrosis was at its peak and then decreased gradually. Immunoprecipitation and Western blot demonstrated that there was enhanced binding of caveolin-1 with eNOS in the process of rat liver cirrhosis. An increase in caveolin-1 expression was detected but the expression of eNOS was lower in cirrhotic tissues than in normal liver tissues. Caveolin-1 protein levels were positively correlated with the degrees of liver fibrosis and the levels of PVP (r=0.967, P < 0.01; r=0.922, P < 0.01, respectively), while NOS catalytic activity was negatively correlated with the degrees of liver fibrosis and levels of PVP (r= 0.973, P < 0.01; r=-0.947, P < 0.01) respectively. CONCLUSIONS: Caveolin-1 upregulation is associated with the development of portal hypertension in liver cirrhosis. Over-expression of caveolin-1 in perisinusoidal cells may promote caveolin-1-eNOS binding and reduce the activity of eNOS.


Subject(s)
Caveolin 1/metabolism , Hypertension, Portal/metabolism , Liver Cirrhosis, Experimental/metabolism , Nitric Oxide Synthase Type III/metabolism , Animals , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...