Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Liver Int ; 39(1): 147-157, 2019 01.
Article in English | MEDLINE | ID: mdl-30178471

ABSTRACT

BACKGROUND & AIMS: Resistance to EGFR-targeted therapy is a major obstacle in hepatocellular carcinoma (HCC) treatment, but its underlying mechanism remains unclear. Autophagy plays a vital role in antitumour treatment. Our previous study suggested that p57 is associated with autophagy and cisplatin resistance. The present study aimed to investigate whether p57 can enhance the sensitivity of HCC cells to Erlotinib (Er)/Cetuximab(C-225) and further explore the potential mechanisms of Er/C-225 resistance. METHODS: HCC cells were transfected with pIRES2-EGFP-p57 and pIRES2-EGFP-nc, accompanied by Er/C-225 treatment. Cell viability was detected by an Annexin apoptosis kit and MTT assay. Xenograft experiments were performed to study the function of p57 in the treatment of Er/C-225 in vivo. The level of autophagy was determined by analysis of the appearance of autophagic vacuoles. Western blotting was used to investigate the potential pathways involved. RESULTS: Up-regulation of p57 decreased the level of Er/C-225-induced autophagy and enhanced the decrease in Er/C-225-induced cell viability. P57 overexpression combined with CQ treatment further enhanced the therapeutic efficiency of Er/C-225. The xenograft experiment verified that p57 up-regulation sensitizes HCC cells to Er/C-225. Moreover, a mechanistic investigation demonstrated that the up-regulation of p57 resulted in a decrease of LC3B-II and beclin-1, and an increase in p-PI3K, p-AKT and p-mTOR protein expressions. CONCLUSIONS: Through activating the PI3K/AKT/mTOR signalling pathway, p57 can reverse Er/C-225-induced autophagy, and thereby increase the therapeutic efficiency of Er/C-225 treatment. Given these results, p57 up-regulation may be applicable as a therapeutic strategy to improve EGFR-targeted therapy in HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy , Carcinoma, Hepatocellular/genetics , Cyclin-Dependent Kinase Inhibitor p57/genetics , ErbB Receptors/antagonists & inhibitors , Liver Neoplasms/genetics , Animals , Apoptosis , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Cell Proliferation , Cetuximab/pharmacology , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Erlotinib Hydrochloride/pharmacology , Humans , Liver Neoplasms/drug therapy , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
2.
Cancer Sci ; 108(10): 1985-1995, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28771881

ABSTRACT

Aberrant expression of microRNAs (miRs) has been shown to play a critical role in the pathogenesis and progression of tumors. microRNA-219-5p (miR-219-5p) has been reported to be abnormally expressed in some types of human tumors. However, the mechanism between miR-219-5p and colorectal cancer (CRC) metastasis remains unclear. In the present study, miR-219-5p was found to be downregulated in CRC tissue compared with matched normal tissue. Through luciferase reporter assay, we demonstrated lymphoid enhancer-binding factor 1 (LEF1) as a direct target of miR-219-5p. Overexpression of miR-219-5p could inhibit motility, migration and invasion of CRC cells, and inhibit epithelial-mesenchymal transition (EMT). Furthermore, silencing LEF1 phenocopied this metastasis-suppressive function. The recovery experiment showed that re-expression of LEF1 rescued this suppressive effect on tumor metastasis and reversed the expression of EMT markers caused by miR-219-5p. Additionally, we demonstrated that miR-219-5p exerted this tumor-suppressive function by blocking activation of the AKT and ERK pathways. Finally, a nude mice experiment showed that miR-219-5p reduced the lung metastasis ability of CRC cells. Taken together, our findings indicate that miR-219-5p inhibits metastasis and EMT of CRC by targeting LEF1 and suppressing the AKT and ERK pathways, which may provide a new antitumor strategy to delay CRC metastasis.


Subject(s)
Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Lung Neoplasms/secondary , Lymphoid Enhancer-Binding Factor 1/genetics , MicroRNAs/genetics , 3' Untranslated Regions , Animals , Caco-2 Cells , Cell Line, Tumor , Colorectal Neoplasms/genetics , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Lung Neoplasms/genetics , MAP Kinase Signaling System , Male , Mice , Mice, Nude , Neoplasm Transplantation
3.
Biomarkers ; 21(3): 272-5, 2016.
Article in English | MEDLINE | ID: mdl-26848614

ABSTRACT

BACKGROUND: Interleukin-27 (IL-27) has been recognized as a pleiotropic cytokine with both pro- and anti-inflammatory properties. PATIENTS AND METHODS: A case-control study was conducted to investigate the possible associations of IL-27 gene polymorphisms with susceptibility to cervical cancer and clinical outcome. RESULTS: Our results suggested that the IL-27 2905T/G was significantly associated with a decreased risk of cervical cancer. Further analysis showed IL-27 2905T/G genotypes were associated with advanced tumor stages of cervical cancer patients. More interestingly, the IL-27 2905T/G genotypes were statistically significantly associated with the survival in cervical cancer patients. CONCLUSION: Our results showed that the IL-27 2905T/G genotypes were associated with decreased the susceptibility and development of cervical cancer in Chinese Han population.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Interleukins/genetics , Uterine Cervical Neoplasms/genetics , Adult , Aged , Alleles , Female , Genotype , Humans , Middle Aged , Polymorphism, Single Nucleotide , Risk Factors , Uterine Cervical Neoplasms/pathology
4.
Tumour Biol ; 35(5): 4107-12, 2014 May.
Article in English | MEDLINE | ID: mdl-24375196

ABSTRACT

Genetic polymorphisms in the Fas/Fas ligand (FasL) gene were proposed to be associated with susceptibility to cervical cancer, but previous studies reported controversial findings. We performed a meta-analysis to assess the associations between Fas/FasL polymorphisms and susceptibility to cervical cancer. We carried out a literature search in PubMed and Embase databases for studies on the associations between Fas/FasL polymorphisms and susceptibility to cervical cancer. The associations were assessed by odds ratio (OR) together with its 95% confidence intervals (CIs). Eleven individual studies with a total of 6,919 subjects were finally included into the meta-analysis. Overall, there was no association between Fas 1377G > A polymorphism and susceptibility to cervical cancer (A vs. G: OR = 0.99, 95% CI 0.88-1.12, P = 0.91; AA vs. GG: OR = 1.00, 95% CI 0.76-1.32, P = 0.99; AA/GA vs. GG: OR = 0.95, 95% CI 0.81-1.12, P = 0.54; AA vs. GG/GA: OR = 1.11, 95% CI 0.85-1.43, P = 0.45). In addition, there was also no association between FasL 844 T > C polymorphism and susceptibility to cervical cancer (C vs. T: OR = 1.12, 95% CI 0.91-1.36, P = 0.28; CC vs. TT: OR = 1.17, 95% CI 0.90-1.51, P = 0.24; CC/TC vs. TT: OR = 1.13, 95% CI 0.92-1.39, P = 0.24; CC vs. TT/TC: OR = 1.11, 95% CI 0.83-1.50, P = 0.47). In subgroup analysis by ethnicity, there were also no associations between Fas/FasL polymorphisms and susceptibility to cervical cancer in Asians and Africans. In conclusion, Fas 1377G > A polymorphism and FasL 844 T > C polymorphism are both not associated with susceptibility to cervical cancer.


Subject(s)
Fas Ligand Protein/genetics , Genetic Predisposition to Disease , Polymorphism, Genetic , Uterine Cervical Neoplasms/genetics , fas Receptor/genetics , Female , Humans , Risk , Uterine Cervical Neoplasms/etiology
5.
Tumour Biol ; 35(12): 11701-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25293518

ABSTRACT

Epidermal growth factor receptor (EGFR) inhibitor treatment is a strategy for cancer therapy. However, innate and acquired resistance is a major obstacle of the efficacy. Autophagy is a self-digesting process in cells, which is considered to be associated with anti-cancer drug resistance. The activation of EGFR can regulate autophagy through multiple signal pathways. EGFR inhibitors can induce autophagy, but the specific function of the induction of autophagy by EGFR inhibitors remains biphasic. On the one hand, autophagy induced by EGFR inhibitors acts as a cytoprotective response in cancer cells, and autophagy inhibitors can enhance the cytotoxic effects of EGFR inhibitors. On the other hand, a high level of autophagy after treatment of EGFR inhibitors can also result in autophagic cell death lacking features of apoptosis, and the combination of EGFR inhibitors with an autophagy inducer might be beneficial. Thus, autophagy regulation represents a promising approach for improving the efficacy of EGFR inhibitors in the treatment of cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , ErbB Receptors/antagonists & inhibitors , Neoplasms/drug therapy , Animals , ErbB Receptors/metabolism , Humans , Molecular Targeted Therapy , Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
6.
World J Surg Oncol ; 11: 5, 2013 Jan 13.
Article in English | MEDLINE | ID: mdl-23311997

ABSTRACT

BACKGROUND: Gastric cancer is a common and highly lethal malignancy in the world, but its pathogenesis remains elusive. In this study, we focus on the biological functions of CDK-associated Cullin1 (CAC1), a novel gene of the cullin family, in gastric cancer, which may help us to further understand the origin of this malignancy. METHODS: The AGS and MGC803 gastric cancer cell lines and the GES-1 gastric mucosa cell line were selected for study. At first, CAC1 expressions of those cell lines were examined by quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) and western blot examinations, then CAC1 small interfering RNA (CAC1-siRNA) were designed and transfected into the AGS cell line with a relatively high level of CAC1. Once CAC1 was silenced, a series of biological characteristics of AGS cells such as cell proliferation, cell cycle, apoptosis, and expressions of apoptosis-related genes (P53, BCL2 and BAX) were determined by MTT, flow cytometry, qRT-PCR and western blot, respectively. RESULTS: CAC1 expression of AGS or MGC803 was much higher than that of GES-1. After CAC1 expression was effectively depressed by RNA interference in AGS cells, significant cell growth inhibition occurred. Furthermore, the proportion of cells treated with CAC1-siRNA increased in the G1 phase and decreased in the S phase, indicative of G1 cell cycle arrest. More importantly, the proportions of early/late apoptosis in AGS cells were enhanced with cis-diaminedichloroplatinum (cisplatin, CDDP) treatment, but to a higher extent with cisplatin plus CAC1-siRNA. Interestingly, BCL2 mRNA copies showed about a 30% decrease in the cisplatin group, but dropped by around 60% in the cisplatin plus CAC1-siRNA group. Conversely, the P53 mRNA expressions obtained nearly a two-fold increase in the cisplatin group, in addition to a five-fold increase in the cisplatin plus CAC1-siRNA group, and the BAX mRNA levels had almost a two- and four-fold augmentation, respectively. Meanwhile, P53, BAX and BCL2 showed the same alteration patterns in western blot examinations. CONCLUSIONS: CAC1 can promote cell proliferation in the AGS gastric cancer cell line. Moreover, it can prevent AGS cells from experiencing cisplatin-induced apoptosis via modulating expressions of P53, BCL2 and BAX.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Cisplatin/pharmacology , Cullin Proteins/physiology , Gastric Mucosa/drug effects , Stomach Neoplasms/pathology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Cell Cycle/drug effects , Cells, Cultured , Cullin Proteins/antagonists & inhibitors , Flow Cytometry , Gastric Mucosa/metabolism , Humans , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism
7.
Oncol Res ; 20(2-3): 113-21, 2012.
Article in English | MEDLINE | ID: mdl-23193917

ABSTRACT

Hepatocellular carcinoma (HCC) is a complex and heterogeneous tumor with several genomic alterations and ranks as the third highest cause of cancer-related death globally. The unresectable or metastatic HCC has very poor prognosis. Although multikinase inhibitor sorafenib can increase the survival of patients with advanced HCC, it is becoming apparent that combination therapies are critical to overcome the complex genomic aberrations in HCC. PTEN, as one of the most commonly inactivated genes in HCC, exerts a wide range of antitumor effects. In this study, we found that PTEN was downregulated in HCC tissues, especially in those tissues with extrahepatic metastasis. And negative PTEN expression cases showed increased proliferation activity. Overexpression of PTEN significantly reduced the proliferation of tumor cells HepG2. In addition, HepG2 cells transfected with PTEN were more sensitive to sorafenib in terms of its ability to inhibit proliferation and to induce apoptosis. Moreover, overexpression of PTEN decreased phosphorylation of MEK, a key downstream effector of RAF/MEK/ERK cascade, and levels of cyclin D1, antiapoptotic Bcl-2, and VEGF. These observations indicated that combination therapies with sorafenib and PTEN overexpression have potential to further improve therapeutic options for HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Benzenesulfonates/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/drug effects , Drug Resistance/drug effects , PTEN Phosphohydrolase/metabolism , Pyridines/pharmacology , Apoptosis/drug effects , Blotting, Western , Carcinoma, Hepatocellular/mortality , Case-Control Studies , Flow Cytometry , Humans , Immunoenzyme Techniques , Liver/cytology , Liver/drug effects , Liver/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Niacinamide/analogs & derivatives , Phenylurea Compounds , Prognosis , Sorafenib , Survival Rate , Tumor Cells, Cultured
8.
BMC Gastroenterol ; 12: 53, 2012 May 28.
Article in English | MEDLINE | ID: mdl-22639890

ABSTRACT

BACKGROUND: Colon cancer is one of the most aggressive human malignancies, with a very poor prognosis. Although it has been suggested that different isoforms of the lymphoid enhancer factor (LEF-1) have opposing biological activities, the biological outcome of aberrant LEF-1 activation in colon cancer is still unclear. The aim of this study was to evaluate the effect of the different LEF-1 phenotypes on the growth of colon carcinoma cell lines. A deeper understanding of these processes might improve the targeted therapies for colon cancer by regulating the expression of LEF-1. METHODS: The role of different isoforms of LEF-1 on the growth of human colon carcinoma cell lines (SW480 and HT-29) was studied using various in vitro and in vivo assays. In vitro proliferation, migration, adhesion and apoptosis of the cells stably transfected of different isoforms of LEF-1 were monitored by MTT assay, carboxyfluorescein diacetate-succinimidyl ester staining, annexin V staining, ECM adhesion assay and transwell assay, respectively. In nude mice, the formation of neovasculature in the tumors formed by our constructed cells was measured by immunohistochemistry. All the data were analyzed using a t test, and data were treated as significant when p < 0.05. RESULTS: Overexpression of truncated LEF-1 (LEF-1-ΔL) in the colon cell lines, SW480 and HT29, inhibited their growth significantly in vitro and in vivo, but the full-length LEF-1 (LEF-1-FL) promoted the proliferation of HT29. Inactivation of Wnt signaling by LEF-1-ΔL reduced the expression of CXCR4 in colon cell lines, which may lead to a decrease in activities such as migration, adhesion and survival. In nude mice, the formation of neovasculature as well as an increase in tumor volume were inhibited by the short isoform of LEF-1. LEF-1-FL, however, caused an increase in all these parameters compared with controls. CONCLUSIONS: These findings suggest that LEF-1 might play an important role in colon carcinogenesis by acting as a regulator. Enhanced expression of LEF-1-FL, which occurs frequently in colon cancer, may be a new target for clinical therapy.


Subject(s)
Cell Proliferation , Colonic Neoplasms/pathology , Colonic Neoplasms/physiopathology , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/physiology , Phenotype , Adenocarcinoma/pathology , Adenocarcinoma/physiopathology , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , HT29 Cells , Humans , In Vitro Techniques , Mice , Mice, Nude , Protein Isoforms , Receptors, CXCR4/physiology , Transplantation, Heterologous , Wnt Proteins/physiology
9.
Int J Colorectal Dis ; 27(3): 287-98, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22065111

ABSTRACT

PURPOSE: Pleiotrophin (PTN) is an important developmental secretory cytokine expressed in many types of cancer and involved in angiogenesis and tumor growth; however, the significance of PTN expression in colorectal cancer (CRC) has not been established. METHODS: Immunohistochemistry, western blot, and enzyme-linked immunosorbent assay were used to detect PTN expression in CRC patients. The relationship between PTN expression and clinicopathological characteristics and survival time was statistically analyzed, and the relationship between PTN and vascular endothelial growth factor (VEGF) in tumor angiogenesis was further analyzed. RESULTS: Of CRC tissues, 74.70% (62/83) stained positive, with a strong positive ratio of 60.24% (50/83). The expression of PTN in CRC tissues was much higher than in normal colorectal tissues. PTN serum levels in CRC patients (mean = 254.59 Ā± 261.76 pg/ml) were significantly higher than those of normal volunteers (mean = 115.23 Ā± 79.53 pg/ml; p < 0.001). PTN expression was related to CRC differentiation and TNM staging. High level of PTN is a predictor of a poor prognosis and high expression of PTN is accompanied by high expression of VEGF in CRC patients. Investigation of the relationship between PTN and VEGF revealed that PTN, through the PTN/RPTPƟ/ƎĀ¶ signaling pathway, increased tyrosine phosphorylation of Ɵ-catenin, leading to an increase in VEGF. CONCLUSIONS: Our study identifies PTN as an essential growth factor for CRC. PTN promotes VEGF expression and cooperates with VEGF in promoting CRC angiogenesis. PTN could serve as a prognostic factor for this cancer. Considering that PTN shows very limited expression in normal tissue, it may represent an attractive new target for CRC therapy.


Subject(s)
Adenocarcinoma/metabolism , Carrier Proteins/metabolism , Colonic Neoplasms/metabolism , Cytokines/metabolism , Rectal Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adenocarcinoma/blood supply , Adenocarcinoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Analysis of Variance , Caco-2 Cells , Carrier Proteins/blood , Carrier Proteins/pharmacology , Child , Colon/metabolism , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Cytokines/blood , Cytokines/pharmacology , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neovascularization, Pathologic/metabolism , Phosphorylation/drug effects , Rectal Neoplasms/blood supply , Rectal Neoplasms/pathology , Rectum/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/drug effects , Young Adult , beta Catenin/drug effects , beta Catenin/metabolism
10.
Carcinogenesis ; 32(12): 1897-904, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22002319

ABSTRACT

p57 is a multifunctional protein involved in the regulation of tumor formation and development; however, the biological role of p57 in the pathogenesis of hepatocellular carcinoma (HCC) is poorly understood. To explore the role of p57 in the development of HCC, we examined p57 messenger RNA (mRNA) and protein levels in HCC tissues and adjacent non-cancerous tissues by immunohistochemistry, real-time polymerase chain reaction and western blot analysis. Moreover, we generated stable p57 knockdown HCC cell lines to investigate the impact of p57 downregulation on the growth and invasion of HCC in vitro and in vivo. Our results showed that p57 mRNA and protein levels were significantly decreased in human HCC tissues. In addition, this reduction in p57 expression was associated with increased tumor size, more advanced TNM stages, the presence of capsule invasion and extrahepatic metastasis and decreased overall survival time. In human HCC cell lines, p57 downregulation increased the expression of cyclin D1 and CDK2 and enhanced the activities of CDK4/cyclin D1 and CDK2/cyclin E complexes, resulting in increased cellular proliferation and growth of xenografts. Furthermore, p57 downregulation accelerated the invasion of HCC cells in vitro and in vivo by controlling the activity of LIMK1. In conclusion, the downregulation of p57 accelerates the growth and invasion of HCC, indicating that p57 is an important tumor suppressor in HCC. Based on these findings, p57 may be a potential target for HCC prevention and therapy.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Division/physiology , Cyclin-Dependent Kinase Inhibitor p57/physiology , Down-Regulation , Liver Neoplasms/pathology , Neoplasm Invasiveness , Adult , Aged , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p57/genetics , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Middle Aged , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
11.
Carcinogenesis ; 31(7): 1211-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20430845

ABSTRACT

Hepatocellular carcinoma (HCC) is a typical hypervascular tumor, and increased levels of vascular endothelial growth factor (VEGF) are associated with progression of HCC. Tumor suppression gene PTEN (phosphatase and tensin homolog deleted on chromosome 10), an important antagonist of the phosphoinositide-3-kinase (PI3K)/adenosine triphosphate-dependent tyrosine kinase (Akt) pathway, is also commonly lost or mutated in HCC. However, the effect of PTEN on VEGF-mediated angiogenesis in HCC remains unknown. To explore this relationship, we expressed a panel of PTEN mutants in human HCC cells with low expression of PTEN (HepG2 cells). Overexpression of PTEN in HepG2 cells resulted in the downregulation of proliferation and migration of cocultured endothelial cells and decreased expression of hypoxia-inducible factor 1 (HIF-1) and VEGF. Similarly, using a nude mouse model, we demonstrated that PTEN decreased expression of HIF-1 and VEGF and suppressed HepG2-induced angiogenesis. This inhibitory effect was not observed in cells expressing a phosphatase-deficient PTEN mutant, suggesting that PTEN inhibits angiogenesis and VEGF through a phosphatase-dependent pathway. Strikingly, reintroducing the C2 domain of PTEN also resulted in a significant decrease in angiogenesis and VEGF expression, although it did not affect Akt phosphorylation or HIF-1 expression. In summary, this study suggests the novel viewpoint that PTEN suppresses angiogenesis and VEGF expression in HCC through both phosphatase-dependent and -independent mechanisms.


Subject(s)
Carcinoma, Hepatocellular/blood supply , Liver Neoplasms/blood supply , Neovascularization, Pathologic/prevention & control , PTEN Phosphohydrolase/physiology , Vascular Endothelial Growth Factor A/genetics , Animals , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , RNA, Messenger/analysis , Vascular Endothelial Growth Factor A/physiology , Xenograft Model Antitumor Assays
12.
Mol Biol Rep ; 37(7): 3407-12, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20012371

ABSTRACT

The anti-proliferative, cytotoxic and apoptogenic activities of delisheng, a Chinese medicinal compound, has been investigated. In this study, the hepatocarcinoma cell line (HepG2) and the liver cell line (L-02) were exposed to delisheng (6.25, 50 and 100 Āµl/ml). Delisheng suppressed the proliferation and viability of normal liver L-02 cells slightly, but strongly inhibited the proliferation and viability of hepatocarcinoma HepG2 cells. The flow cytometric analysis of HepG2 cells demonstrated that delisheng primarily arrested the HepG2 cells at the G1 phase of the cell cycle. Annexin V-FITC/PI staining corroborates the apoptogenic nature of delisheng on HepG2 cells. The anti-proliferative and pro-apoptotic effect of delisheng in HepG2 cells was associated with changes in the Bcl-2/Bax ratio and the induction of caspase-mediated apoptosis. Upregulation of DR5 expression was observed in HepG2 cells after treatment with delisheng. The findings from the present study suggest that delisheng has selective cytotoxic activities against HepG2 cells. Delisheng triggered time- and dose-dependent apoptosis in HepG2 cells by activating the mitochondria-mediated and death receptor-mediated apoptotic pathways.


Subject(s)
Apoptosis/drug effects , Drugs, Chinese Herbal/pharmacology , Signal Transduction/drug effects , Annexin A5/metabolism , Blotting, Western , Caspases/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Flow Cytometry , Fluorescein-5-isothiocyanate/metabolism , Hep G2 Cells , Humans , Propidium/metabolism , Receptors, Death Domain/metabolism , bcl-2-Associated X Protein/metabolism
13.
J Exp Clin Cancer Res ; 39(1): 284, 2020 Dec 14.
Article in English | MEDLINE | ID: mdl-33317597

ABSTRACT

The enthusiasm for immune checkpoint inhibitors (ICIs), an efficient tumor treatment model different from traditional treatment, is based on their unprecedented antitumor effect, but the occurrence of immune-related adverse events (irAEs) is an obstacle to the prospect of ICI treatment. IrAEs are a discrete toxicity caused by the nonspecific activation of the immune system and can affect almost all tissues and organs. Currently, research on biomarkers mainly focuses on the gastrointestinal tract, endocrine system, skin and lung. Several potential hypotheses concentrate on the overactivation of the immune system, excessive release of inflammatory cytokines, elevated levels of pre-existing autoantibodies, and presence of common antigens between tumors and normal tissues. This review lists the current biomarkers that might predict irAEs and their possible mechanisms for both nonspecific and organ-specific biomarkers. However, the prediction of irAEs remains a major clinical challenge to screen and identify patients who are susceptible to irAEs and likely to benefit from ICIs.


Subject(s)
Biomarkers/metabolism , Drug-Related Side Effects and Adverse Reactions/diagnosis , Immune Checkpoint Inhibitors/adverse effects , Neoplasms/drug therapy , Drug-Related Side Effects and Adverse Reactions/etiology , Drug-Related Side Effects and Adverse Reactions/metabolism , Humans , Neoplasms/immunology , Neoplasms/pathology
14.
Biochem Biophys Res Commun ; 388(1): 67-72, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19643084

ABSTRACT

The function of T cells and B cells is to recognize specific "non-self" antigens, during a process known as antigen presentation. Once they have identified an invader, the cells generate specific responses that are tailored to maximally eliminate specific pathogens or pathogen-infected cells. Endothelial cells (ECs) can trigger the activation of T cells through their class I and class II MHC molecules. In this study, we examined the effect of ECs on the proliferation of lymphocytes. We report that the proliferation of T and B cells can be improved by interaction with ECs. LEF-1 is one of the main molecular mediators in this process, and the inhibition of LEF-1 induces apoptosis. These results suggest that LEF-1 modulates positively the proliferation of lymphocytes induced by their interaction with ECs.


Subject(s)
Endothelium, Vascular/immunology , Lymphocyte Activation , Lymphocytes/immunology , Lymphoid Enhancer-Binding Factor 1/metabolism , Wnt Proteins/metabolism , Animals , Apoptosis , Cell Proliferation , Down-Regulation , Endothelium, Vascular/cytology , Mice
15.
Int J Neuropsychopharmacol ; 12(8): 1089-96, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19272208

ABSTRACT

Recent data from several studies suggest that oxidative stress is involved in the biochemical mechanisms that underlie neuropsychiatric disorders. The present study was designed to investigate oxidative stress status in depressive patients with gastric adenocarcinoma (GA) at TNM stage III. Oxidative stress, depression and expression of specific genes were monitored during a pretreatment period. Serum total antioxidant capacity, catalase, superoxide dismutase concentrations, and antisuperoxide anion capacity (A-ASC) were significantly decreased in depressive patients compared to control subjects, whereas serum malondialdehyde (MDA) levels were significantly increased. Importantly, the formation of 8-hydroxy-deoxyguanosine (8-OHdG) accumulated. Furthermore, SYBR Green real-time PCR revealed that the expression levels of human oxoguanine glycosylase 1 and APEX nuclease 1 (APEX1) were increased in depressive patients. Pearson correlation analysis revealed that depression was positively correlated with SAS, SCL-90, MDA, 8-OHdG and APEX1, but negatively correlated with A-ASC. Thus, this study confirms oxidative imbalance in depressive patients with GA, and oxidative stress may play a role in the onset and exacerbation of depression.


Subject(s)
Adenocarcinoma/complications , Depression/complications , Oxidative Stress/physiology , Stomach Neoplasms/complications , 8-Hydroxy-2'-Deoxyguanosine , Adult , Aged , Analysis of Variance , Antioxidants/metabolism , Catalase/blood , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Male , Malondialdehyde/metabolism , Middle Aged , Superoxide Dismutase/blood
16.
Oncol Rep ; 21(4): 925-31, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19287990

ABSTRACT

LKB1 encodes a serine/threonine kinase generally inactivated in human lung cancers, which mediates cancer cell proliferation, migration and differentiation, but its biological function has not been completely elucidated. In this study, we demonstrated that LKB1 was associated with a substantial reduction of c-myc expression by using an inducible LKB1 expression system in the LKB1-null lung cell line A549. Nevertheless, the reduction of the c-Myc gene expression was not accompanied by corresponding reduction of mRNAs but protein, which can be abrogated by a proteosome inhibitor (MG132), suggesting that the reduction was associated with their increased degradation rather than transcriptional controls. Our results implied that the expression of c-Myc protein decreased by LKB1 in transfected cells may be a contributory factor in the process of cell proliferation. Overexpression of the LKB1 gene could inhibit the activation of ERK1/2 and STAT3 signaling pathways involved in the cell proliferation. Thus, LKB1-induced functional operation on c-Myc in promoting cell proliferation may occur in a novel mechanism, which may be regulated by ERK1/2 and/or STAT3 signal pathways in human lung carcinoma cells. Furthermore, our results give some insights into the understanding of how LKB1 inactivation contributes to lung carcinogenesis and emphasizes the central role played by LKB1 in lung cancer development.


Subject(s)
Lung Neoplasms/prevention & control , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-myc/metabolism , STAT3 Transcription Factor/physiology , AMP-Activated Protein Kinase Kinases , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Lung Neoplasms/genetics , MAP Kinase Signaling System , Protein Serine-Threonine Kinases/genetics
17.
J Psychosom Res ; 66(3): 259-66, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19232240

ABSTRACT

OBJECTIVES: This study investigated the connection among the oxidative stress, depression and expression of specific genes involved in DNA-damage signaling pathways in patients with colorectal carcinoma (CRC). METHODS: A unique Dukes'C subset of patients with newly diagnosed colorectal adenocarcinoma were assessed using the Hamilton Depression Rating Scale (HAMD), Zung Self-rating Depression Scale (SDS), Zung Self-rating Anxiety Scale (SAS), Symptom Checklist 90 (SCL-90) and other multiple-item questionnaires. Oxidative-stress-related parameters in sera and the expression of genes were monitored during a pretreatment period. RESULTS: Eighty-two eligibility cases were divided into 2 groups based on an HAMD score cutoff of 20: the mean score was 28.29 in Group A (depression, n=52) and 16.50 in Group B (nondepression, n=30). The serum total antioxidant capacity, catalase, and superoxide dismutase concentrations were lower in Group A, whereas those of nitric oxide and malondialdehyde were higher in Group A. Importantly, the 8-hydroxy-deoxyguanosine level was higher in Group A than in Group B (P<.05). Microarray analysis revealed that the expressions of p34, PA26, and ABL were higher in Group A, whereas those of HRAD51, CR6, and XRCC3 were higher in Group B. CONCLUSION: Oxidative stress is capable of causing neuronal toxicity via lipid peroxidation, DNA damage, and abnormalities of gene expression, and therefore is a possible pathogenic mechanism underlying depression in patients with CRC.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , DNA Damage/genetics , Depressive Disorder/genetics , Gene Expression Regulation, Neoplastic/genetics , Oxidative Stress/genetics , Signal Transduction/genetics , 8-Hydroxy-2'-Deoxyguanosine/analogs & derivatives , Adenocarcinoma/pathology , Adenocarcinoma/psychology , Adult , Aged , Colorectal Neoplasms/pathology , Colorectal Neoplasms/psychology , Depressive Disorder/complications , Depressive Disorder/psychology , Female , Guanine/analogs & derivatives , Guanine/blood , Humans , Male , Middle Aged , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Oxidative Stress/physiology , Personality Inventory/statistics & numerical data , Psychometrics , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors
18.
Cancer Manag Res ; 11: 4283-4295, 2019.
Article in English | MEDLINE | ID: mdl-31190994

ABSTRACT

Background: The prognostic and clinicopathological role of pretreatment thrombocytosis in cancer has been widely studied, but conclusions in endometrial cancer (EnCa) remain controversial. Therefore, we conducted a meta-analysis to assess the pathologic and prognostic impacts of pretreatment thrombocytosis in patients with EnCa. Methods: We searched PubMed, Embase, SpringerLink, ScienceDirect and China National Knowledge Infrastructure databases. Pooled HR or OR with their 95% CIs were applied to assess the association of pretreatment thrombocytosis with survival outcomes and clinical parameters of EnCa patients. Results: In total, 10 studies containing 2,995 cases of EnCa met the criteria. The results suggested that pretreatment thrombocytosis was significantly associated with high International Federation of Gynecology and Obstetrics (FIGO) stage (pooled OR 3.45, 95% CI 1.68-7.08, P=0.001), poor tumor differentiation (pooled OR 2.00, 95% CI 1.22-3.29, P=0.006), lymph-vascular space invasion (pooled OR 2.04, 95% CI 1.35-3.07, P=0.001); myometrial invasion (pooled OR 2.14, 95% CI 1.39-3.32, P=0.001); cervical involvement (pooled OR 2.54, 95% CI 1.56-4.15, P=0.000) and lymph node metastasis (OR 3.15, 95% CI 1.71-5.80, P=0.001). No significant difference existed between pretreatment thrombocytosis and overall survival (P=0.012), cancer/disease-specific survival (P=0.07) or disease-free survival (P=0.25). Conclusion: pretreatment thrombocytosis was associated with advanced clinicopathological features in patients with EnCa, which may serve as a potential therapeutic target for EnCa.

19.
Oncol Rep ; 20(6): 1465-71, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19020729

ABSTRACT

Mammalian cells grow in three-dimensions (3-D) in vivo. Commonly used two-dimensional (2-D) cell cultures are inadequate to recreate the biological microenvironment of tumor cells. The potentially different outcomes from 2-D and 3-D culture systems may have a significant impact on the relevance of experimental findings. The purpose of this study was to characterize the human hepatoma cell line HepG2 in 2-D and 3-D cultures. HepG2 cells in 2-D and 3-D cultures were treated with cisplatin, 5-fluorouracil, and adriamycin and were analyzed by scanning electron microscopy and transmission electron microscopy. Cell cycle progression and apoptosis were detected by flow cytometry. Inhibition of cell proliferation was quantified by MTT assay. The expression of E-cadherin, CD44v6, VEGF, KDR, endostatin, Bax, and cytochrome-c were analyzed by immunohistochemical (IHC) staining. As compared to the 2-D monolayer culture, the 3-D multicellular spheroids (MCS) of HepG2 cells featured a greater fraction of cells in G1 phase and were organized with more abundant cell-cell adhesion. In addition, cells in MCS were significantly less apoptotic in maintenance culture media and were more resistant to drug-induced apoptosis. E-cadherin, CD44v6, VEGF, KDR, endostatin, and cytochrome-c levels were increased in MCS as compared to 2-D cell cultures. In conclusion, MCS conferred differentiated phenotypes including increased cell-cell adhesion and G1 phase cell cycle arrest, enhanced cellular resistance to apoptosis, and upregulated angiogenic potential. Based on our data, a multicellular morphological hierarchy may sustain the growth/survival advantages of cancer cells in vivo. Therefore, a 3-D culture system should be the preferred technique for cancer biology investigation.


Subject(s)
Spheroids, Cellular/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Cadherins/biosynthesis , Cell Line, Tumor , Cell Survival , Cisplatin/pharmacology , Doxorubicin/pharmacology , Fluorouracil/pharmacology , Humans , Hyaluronan Receptors/biosynthesis , In Vitro Techniques , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/biosynthesis
20.
Zhonghua Zhong Liu Za Zhi ; 30(6): 465-8, 2008 Jun.
Article in Zh | MEDLINE | ID: mdl-19024525

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the efficacy, toxicity and safety of doxorubicin combined with domestically produced docetaxel versus with taxotere, and to investigate whether these two regimens result in similar outcomes in the treatment for non-small-cell lung cancer (NSCLC) patients who failed previous platinum-based chemotherapy. METHODS: Eighty-eight NSCLC patients were enrolled into this clinical phase II trial. The patients randomly received either domestic docetaxel (study arm) or taxotere (control arm) at a dose of 70 mg/m2 on D2, while doxorubicin at a dose of 40 mg/m2 on D1 was administered in both groups. It was repeated every 3 weeks, totally for three cycles. No granulocyte colony-stimulating factor was used to prevent granulocytopenia. The response rate and toxicity were evaluated using World Health Organization toxicity scale and Karnofsky performance status scale. RESULTS: Of the 88 patients, 81 were evaluable in terms of efficacy. There was no complete responder in this series. The response rate (RR) was 17.1% in the study arm versus 7.5% in the control arm, and the clinical benefit rate (CBR) was 80.5% in the study group versus 72.5% in the control group. The most frequent grade 3 or 4 toxicities were neutropenia, leucopenia and gastrointestinal symptoms. Other toxicities such as alopecia and vomiting were mild and generally well tolerated. No fluid retention was noticed. CONCLUSION: The administration of doxorubicin 40 mg/m2 on D1 combined with domestic docetaxel 70 mg/m2 on D2 is proved to be as effective and tolerable as with taxotere. The domestic drug docetaxel may be considered as an alternative for patients with non-small-cell lung cancer who failed previous platinum-based chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Non-Small-Cell Lung/pathology , Docetaxel , Doxorubicin/administration & dosage , Female , Humans , Leukopenia/chemically induced , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Neutropenia/chemically induced , Remission Induction , Salvage Therapy , Taxoids/administration & dosage , Treatment Failure , Vomiting/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL