Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Cancer Sci ; 112(3): 1060-1074, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33340431

ABSTRACT

Metastasis-associated protein 2 (MTA2) is frequently amplified in many types of cancers; however, the role and underlying molecular mechanism of MTA2 in esophageal squamous cell carcinoma (ESCC) remain unknown. Here, we reported that MTA2 is highly expressed in ESCC tissue and cells, and is closely related to the malignant characteristics and poor prognosis of patients with ESCC. Through in vitro and in vivo experiments, we demonstrated that MTA2 significantly promoted ESCC growth, metastasis, and epithelial-mesenchymal transition (EMT) progression. This integrative analysis combined with expression microarray showed that MTA2 could interact with eukaryotic initiation factor 4E (EIF4E), which positively regulates the expression of Twist, known as a master regulator of EMT. Moreover, the results of chromatin immunoprecipitation revealed that MTA2 was recruited to the E-cadherin promoter by Twist, which reduced the acetylation level of the promoter region and thus inhibited expression of E-cadherin, and subsequently promoted the aggressive progression of ESCC. Collectively, our study provided novel evidence that MTA2 plays an aggressive role in ESCC metastasis by a novel EIF4E-Twist positive feedback loop, which may provide a potential therapeutic target for the management of ESCC.


Subject(s)
Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Eukaryotic Initiation Factor-4E/metabolism , Histone Deacetylases/metabolism , Nuclear Proteins/genetics , Repressor Proteins/metabolism , Twist-Related Protein 1/genetics , Animals , Antigens, CD/genetics , Cadherins/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Neoplasms/surgery , Esophageal Squamous Cell Carcinoma/mortality , Esophageal Squamous Cell Carcinoma/secondary , Esophageal Squamous Cell Carcinoma/surgery , Esophagectomy , Esophagus/pathology , Esophagus/surgery , Eukaryotic Initiation Factor-4E/genetics , Feedback, Physiological , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histone Deacetylases/genetics , Humans , Kaplan-Meier Estimate , Male , Mice , Middle Aged , Nuclear Proteins/metabolism , Prognosis , Promoter Regions, Genetic , Repressor Proteins/genetics , Twist-Related Protein 1/metabolism , Xenograft Model Antitumor Assays
2.
Mol Carcinog ; 58(6): 1033-1045, 2019 06.
Article in English | MEDLINE | ID: mdl-30737960

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a member of the tumor necrosis factor family, induces apoptosis in a variety of cancer cells. However, gastric cancer (GC) cells are insensitive to TRAIL usually. In the previous study, we showed that Periplocin could induce apoptosis in GC cells via the activation of ERK1/2-EGR1 pathway. In the present study, we have shown that the combination of Periplocin and TRAIL had a greater inhibitory effect on gastric cancer cell viability in vitro and in vivo than Periplocin or TRAIL alone. Through upregulating the expression of DR4 and DR5 at transcriptional and protein levels, Periplocin enhanced the sensitivity of gastric cancer cells to TRAIL. Furthermore, enhanced activity of ERK1/2-EGR1 pathway was responsible for upregulating of DR4 and DR5 uponPeriplocin treatment, subsequently reducing the expression of Mcl-1 and Bcl2 and activating Bid and caspase-3/8. Collectively, these data implied that Periplocin might act as a sensitizer of TRAIL and could be a potential strategy for the treatment of GC.


Subject(s)
Drug Resistance, Neoplasm/drug effects , MAP Kinase Signaling System/drug effects , Saponins/administration & dosage , Stomach Neoplasms/drug therapy , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Saponins/pharmacology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Up-Regulation , Xenograft Model Antitumor Assays
3.
Mutagenesis ; 34(4): 307-313, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31165868

ABSTRACT

Baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5) is an inhibitor of apoptosis proteins and plays a key role in apoptosis or programmed cell death. In the present study, we evaluated the effect of BIRC5 gene polymorphisms on the risk of developing oesophageal squamous cell carcinoma (ESCC) and patients' outcomes in a high-incidence population from northern China. A population-based case-control study was performed in 597 ESCC patients and 597 control subjects.Survival data were available for 211 patients who received platinum-based chemotherapy after surgery. Five polymorphisms (-31 C>G, -241 C>T, -625 G>C, -644 T>C and -1547 A>G) in the promoter of the BIRC5 gene were genotyped by the polymerase chain reaction-ligase detection reaction (PCR-LDR) method. Compared with the -31 CC genotype, the -31 CG/GG genotype of -31 C>G single nucleotide polymorphism (SNP) was associated with a significant elevated risk of ESCC [adjusted odds ratio (OR) = 1.40, 95% confidence interval (CI) = 1.07-1.84]. Interestingly, this association was stronger among females, younger patients and non-smokers in stratified analyses (adjusted OR = 1.72, 95% CI = 1.07-2.75; adjusted OR = 1.61, 95% CI = 1.10-2.36; adjusted OR = 1.80, 95% CI = 1.26-2.58, respectively]. Survival analyses showed that the T allele of -241 C>T SNP was associated with poor prognosis [hazard ratio (HR) = 2.99, 95% CI = 1.09-8.19) and that the C allele of -625 G>C SNP was associated with good prognosis (HR = 0.62, 95% CI = 0.38-0.99) in ESCC patients. The -31 C>G polymorphism may be involved in the development of ESCC, and the -241 C>T and -625 G>C polymorphisms may be useful prognostic markers for ESCC.


Subject(s)
Biomarkers, Tumor , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/mortality , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Survivin/genetics , Adult , Aged , Alleles , Esophageal Squamous Cell Carcinoma/pathology , Female , Gene Frequency , Genotype , Haplotypes , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Odds Ratio , Prognosis
4.
Cell Physiol Biochem ; 45(6): 2471-2482, 2018.
Article in English | MEDLINE | ID: mdl-29554660

ABSTRACT

BACKGROUND/AIMS: Small nucleolar RNAs (snoRNAs) play an important role in carcinogenesis. In this study, we identified a C/D box snoRNA, snord105b, and further investigated the function and mechanism of the snord105b in gastric cancer (GC). METHODS: The expression level of snord105b in GC tissures, sera and cell lines were detected by qRT-PCR. Cell viability was assessed using MTS assay. Transwell and wound healing assay were performed to evaluate migration and invasion, and protein expression was examined by western blotting. ChIRP and MS analysis was used to seek for the special binding protein of snord105b. RESULTS: The snord105b was upregulated and associated with tumor size, differentiation, and pathological stage in GC. Snord105b affected proliferation, migration and invasion in multiple GC cell lines. The oncoqenic activity of snord105b was also confirmed with in vivo data. Mechanistically, snord105b specifically bound to ALDOA and affected C-myc, which plays a key role in carcinogenesis and tumor development. CONCLUSION: Snord105b appears to be a novel oncogene and is clinically and functionally involved in the development of GC. Targeting snord105b and its pathway may provide new biomarkers or potential treatments for patients with GC.


Subject(s)
Carcinogenesis/genetics , Fructose-Bisphosphate Aldolase/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Nucleolar/metabolism , Signal Transduction , Stomach Neoplasms/genetics , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Female , Gastric Mucosa/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , RNA, Small Nucleolar/genetics , Stomach/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Up-Regulation
5.
Cell Biochem Funct ; 36(8): 398-407, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30484863

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is a common malignancy without effective therapy. Histone deacetylase inhibitors (HDACIs) have been demonstrated as an emerging class of anticancer drugs for a range of haematological and solid tumours. However, the effect of HDACIs has not yet been investigated on ESCC cells. In this study, HDACIs were initially considered to have anticancer activity for ESCC, due to the high expression of HDAC genes in ESCC cell lines by analysing expression data of 27 ESCC cell lines from the Broad-Novartis Cancer Cell Line Encyclopedia. Next, we used five ESCC cell lines and one normal immortalized esophageal epithelial cell line to screen three HDACIs, panobinostat (LBH589), vorinostat (SAHA), and trichostatin A (TSA), for the ability to inhibit growth. Here, we report that LBH589 more effectively suppressed cell proliferation of ESCC cell lines, in a dose-dependent manner, than TSA and SAHA, as well as had lower toxicity against the SHEE normal immortalized esophageal epithelial cell line. Further experiments indicated that LBH589 treatment significantly inhibited TP53 (mutated TP53) expression, both at the mRNA and protein level, and simultaneously increased p21 and decreased cyclin D1 expression. Taken together, we propose that LBH589 inhibits ESCC cell proliferation mainly through inducing cell cycle arrest by increasing p21 and decreasing cyclin D1 in a p53-independent manner. SIGNIFICANCE OF THE STUDY: In this study, the antitumor activity of HDACIs LBH589, SAHA, and TSA on ESCC was characterized, with LBH589 displaying the most potent anti-proliferative activity while not harming normal immortalized esophageal epithelial cells. Furthermore, we propose that LBH589 exerts its anti-proliferative effect by inducing cell cycle arrest. The ability to specifically target cancer cells indicates therapeutic potential for use of LBH589 in the treatment of ESCC.


Subject(s)
Cell Cycle Checkpoints/drug effects , Histone Deacetylase Inhibitors/pharmacology , Panobinostat/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation/drug effects , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Humans , Hydroxamic Acids/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects
6.
J Biomed Sci ; 24(1): 6, 2017 Jan 11.
Article in English | MEDLINE | ID: mdl-28077137

ABSTRACT

BACKGROUND: The DACT (Dishevelled-associated antagonist of ß-catenin) family of scaffold proteins may play important roles in tumorigenesis. However, the epigenetic changes of DACT1, 2, 3 and their effect on esophageal squamous cell carcinoma (ESCC) have not been investigated so far. The aim of this study was to investigate the promoter methylation and expression of DACT family, in order to elucidate more information on the role of DACT with regard to the progression and prognosis of ESCC. METHODS: MSP and BGS methods were respectively applied to examine the methylation status of DACT; RT-PCR, Western blot and immunohistochemistry methods were respectively used to determine the mRNA and protein expression of DACT; MTT, Colony-formation and Wound-healing assay were performed to assess the effect of DACT1 and DACT2 on proliferation and migration of esophageal cancer cells. RESULTS: Frequent reduced expression of DACT1, DACT2 and DACT3 were found in esophageal cancer cell lines and the expression levels of DACT1 and DACT2 were reversed by 5-Aza-Dc. Decreased mRNA and protein expression of DACT1 and DACT2 were observed in ESCC tumor tissues and were associated with the methylation status of transcription start site (TSS) region. The hypermethylation of CpG islands (CGI) shore region in DACT1 was observed both in tumor and corresponding adjacent tissues but wasn't related to the transcriptional inhibition of DACT1. The methylation status of TSS region in DACT1 and DACT2 and the protein expression of DACT2 were independently associated with ESCC patients' prognosis. CONCLUSIONS: The TSS region hypermethylation may be one of the main mechanisms for reduced expression of DACT1 and DACT2 in ESCC. The simultaneous methylation of DACT1 and DACT2 may play important roles in progression of ESCC and may serve as prognostic methylation biomarkers for ESCC patients.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Carrier Proteins/metabolism , Esophageal Neoplasms/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Aged , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/pathology , Female , Humans , Male , Methylation , Middle Aged , Prognosis
7.
Cell Physiol Biochem ; 38(5): 1939-51, 2016.
Article in English | MEDLINE | ID: mdl-27160973

ABSTRACT

BACKGROUND/AIMS: Periplocin is extracted from the traditional herbal medicine cortex periplocae, which has been reported to suppress the growth of cancer cells. However, little is known about its effect on gastric cancer cells. METHODS: Gastric cancer cells were treated with periplocin, and cell viability was assessed using MTS assay. Flow cytometry and TUNEL staining were performed to evaluate apoptosis, and protein expression was examined by western blotting. Microarray analysis was used to screen for changes in related genes. RESULTS: We found that periplocin had an inhibitory effect on gastric cancer cell viability in a dose-dependent manner. Periplocin inhibited cell viability via the ERK1/2-EGR1 pathway to induce apoptosis. Periplocin also inhibited the growth of tumor xenografts and induced apoptosis in vivo. CONCLUSION: Our results show that periplocin inhibits the proliferation of gastric cancer cells and induces apoptosis in vitro and in vivo, indicating its potential to be used as an antitumor drug.


Subject(s)
Apocynaceae/chemistry , Apoptosis/drug effects , Saponins/pharmacology , Signal Transduction/drug effects , Animals , Apocynaceae/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Early Growth Response Protein 1/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Plant Extracts/chemistry , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transplantation, Heterologous
8.
Cell Physiol Biochem ; 38(6): 2247-60, 2016.
Article in English | MEDLINE | ID: mdl-27188168

ABSTRACT

BACKGROUND/AIMS: Due to its antitumor and gastroprotective properties, cochinchina momordica seed (CMS), has been widely used to treat cancer patients in Asia. Our previous reports have shown that CMS is able to induce the differentiation of B16-F1 melanoma cells. However, its functional component and mechanism remain unclear and are addressed in this study. METHODS AND RESULTS: CMSP (p-hydroxycinnamaldehyde isolated from CMS) inhibited the proliferation, migration and invasiveness of B16-F1 cells both in vivo and in vitro. CMSP also induced the differentiation of B16-F1 cells, as characterized by dendrite-like outgrowth, increased melanogenesis and enhanced tyrosinase activity. Furthermore, CMSP treatment reduced the level of malignant markers of melanoma, specifically S-100B and melanoma-derived growth regulatory protein precursor (MIA), in a concentration-dependent manner. According to a western blot analysis, B16-F1 cells treated with CMSP exhibited a sustained increase in p-P38 and decreased activities of ERK and JNK. Our data further indicated that the downregulation of GTP-RhoA, which was mediated by increased cAMP release, was involved in CMSP-induced changes in MAPK, while LPA (Lysophosphatidic acid) partially reversed CMSP-induced B16 cell differentiation. CONCLUSION: These results demonstrated that CMSP-induced differentiation of B16F1 cells may occur through the RhoA-MAPK axis, which suggests a new potential strategy for melanoma treatment.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Differentiation/drug effects , Cinnamates/pharmacology , Melanoma, Experimental/drug therapy , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/drug effects , rhoA GTP-Binding Protein/metabolism , Animals , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Cinnamates/chemistry , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Momordica/chemistry , Monophenol Monooxygenase/metabolism , Seeds/chemistry
9.
Tumour Biol ; 37(3): 2961-71, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26411671

ABSTRACT

The coinhibitory molecules, B7-H3 and B7-H4, have shown negative regulation in T cell activation and tumor-associated macrophage (TAM) polarization in tumor-specific immunity. Here, we investigated the expression of B7-H3 and B7-H4 in human and murine esophageal squamous cell carcinoma (ESCC) tissues to define their clinical significance and mechanism in a tumor microenvironment. In the present study, B7-H3 and B7-H4 were expressed in 90.6 and 92.7 % samples, respectively. High B7-H3 and B7-H4 expression was associated with advanced TNM stage and lymph node metastasis (p < 0.05, respectively). Patients with both B7-H3 and B7-H4 high-expressed tumors had the poorest prognosis (26.7 months), whereas those with both low-expressed tumors had the best survival (56.7 months). B7-H3 and B7-H4 expression were inclined to be positively related to the infiltration intensity of Treg cells and TAMs (p < 0.05, respectively), and B7-H3 expression is negatively associated with the intensity of CD8(+) T cells (p < 0.05). In 4-nitroquinoline 1-oxide (4-NQO)-induced murine models, high B7-H3 expression could only be detected at carcinoma stage, but abnormal B7-H4 expression appeared a little earlier at dysplasia stage. In vitro studies revealed that knockdown of B7-H3 on tumor cells suppressed ESCC cell migration and invasion, while knockdown of B7-H4 could inhibit ESCC cell growth. Overall, B7-H3 and B7-H4 are involved in ESCC progression and development and their coexpression could be valuable prognostic indicators. Interference of these negative regulatory molecules might be a new strategy for treating ESCC.


Subject(s)
B7 Antigens/physiology , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , V-Set Domain-Containing T-Cell Activation Inhibitor 1/physiology , 4-Nitroquinoline-1-oxide/toxicity , Adult , Aged , Animals , B7 Antigens/analysis , Carcinoma, Squamous Cell/chemically induced , Cell Line, Tumor , Esophageal Neoplasms/chemically induced , Esophageal Squamous Cell Carcinoma , Female , Humans , Lymphocytes, Tumor-Infiltrating/physiology , Mice , Mice, Inbred C57BL , Middle Aged , Quinolones/toxicity , V-Set Domain-Containing T-Cell Activation Inhibitor 1/analysis
10.
Tumour Biol ; 37(11): 14939-14947, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27644254

ABSTRACT

Our previous studies have shown that the expression level of B7 homolog 3 (B7-H3) was correlated with clinical staging and prognosis of osteosarcoma (OS) patients, and its silencing inhibited the proliferation and invasion of OS cells in vitro. However, its overexpression mechanism behind was far from elucidated. On the basis of bioinformatics and the preliminary screening data, we hypothesized that miR-124 might play an important role in OS development and as a lead candidate for modulating B7-H3 expression. In this study, we found that miR-124 was downregulated significantly in OS tumor tissue, compared to normal adjacent tissues (NATs). Lower miR-124 expression levels were associated with advanced Ennecking stage, lower tumor differentiation, and common pulmonary metastasis. The 5-year overall survival rate in the miR-124 upregulated group was 61.5 %, while with low miR-124 expression, only 11.8 % survived. Further studies in vitro showed that B7-H3 was a direct target of miR-124. Overexpression of miR-124 decreased B7-H3 mRNA and protein level and inhibited B7-H3 3'-UTR reporter activity. Treatment of OS cells with miR-124 mimics induced the inhibition of cell growth and invasion in vitro, which could be abrogated by transfected by B7-H3 expression vector. Our findings highlight the potential application of miR-124 as a novel onco-miRNA in OS, and its oncogenic effects are mediated chiefly through downregulation of B7-H3, thus suggesting a model for identifying miR-124 that can be exploited to improve the therapeutic potential efficacy of mAb targeting to B7-H3.


Subject(s)
B7 Antigens/biosynthesis , Bone Neoplasms/pathology , Cell Movement/genetics , Cell Proliferation/genetics , MicroRNAs/genetics , Osteosarcoma/pathology , Adult , B7 Antigens/genetics , Bone Neoplasms/genetics , Bone Neoplasms/mortality , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/secondary , Male , Neoplasm Invasiveness/genetics , Oncogenes/genetics , Osteosarcoma/genetics , Osteosarcoma/mortality , Young Adult
11.
Zhongguo Zhong Yao Za Zhi ; 40(6): 1207-11, 2015 Mar.
Article in Zh | MEDLINE | ID: mdl-26226772

ABSTRACT

To investigate the effect of Tanreqing injection on immune activity of peripheral blood lymphocytes of patients with lung cancer. The peripheral blood lymphocytes of patients with lung cancer and healthy persons were separated by the density gradient centrifugation method for subsequent experiments, with those from healthy persons as the positive control. The effect of Tanreqing injection on stimulating the proliferation of lymphocytes with phytohemagglutinin (PHA) was determined by MTT method. The effect of Tanreqing injection on the lymphocyte secretions of IFN-γ and TNF-α and the subset ratio of lymphocytes cultured separately or with Tanreqing injection of different concentrations were examined by ELISA and flow cytometry (FCM) respectively. In addition, the LDH release assay was used to detect the cytotoxicity of cytotoxic T cells (CTL) and natural killer cells (NK). According to the findings, all of immunological indexes of lymphocytes from patients with lung cancer were weaker than that of healthy persons, but with the obvious increases in proliferation activity and IFN-γ and TNF-α secretions of lymphocytes co-cultured with Tanreqing Injection (P < 0.05). Among lymphocyte subsets co-cultured with Tanreqing Injection, CD3+, CD3+ CD4+ and CD3- CD16 + 56+ cell ratios notably increased, whereas CD4+ CD25+ Treg cell ratio obviously decreased (P < 0.05). In the meantime, Tanreqing injection can markedly promote the cytotoxicities of CTL and NK (P < 0.05). In conclusion, Tanreqing injection shows a significant effect in promoting the immune activity of lymphocytes from patients with lung cancer and their anti-tumor immunity.


Subject(s)
Drugs, Chinese Herbal/administration & dosage , Killer Cells, Natural/immunology , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Cell Proliferation/drug effects , Cells, Cultured , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Killer Cells, Natural/drug effects , Lung Neoplasms/genetics , Lung Neoplasms/physiopathology , T-Lymphocytes, Cytotoxic/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
12.
Int J Cancer ; 134(12): 2764-71, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24013874

ABSTRACT

B7-H3 (CD276), a newly identified member of the B7 family of molecules, is often induced in human tumors and its overexpression is closely correlated with survival, prognosis or tumor grade. Although cancer immunotherapy has not been completely translated into clinical successes, interest has been further enhanced by the realization of these costimulatory molecules' potential as targets to modulate clinical immune responses. Despite ample evidence implicating B7-H3 in tumor immune escape, a steady flow of reports have suggested that it may also have antitumor effects under certain circumstances. The safety and efficacy of targeting B7-H3 with a monoclonal antibody for the treatment of advanced-stage central nervous system cancer in children has been proven, making B7-H3 an attractive therapeutic target for this kind of tumor. In addition, B7-H3 was shown to promote invasion and accelerate carcinogenesis in tumor progression according to its nonimmunological regulatory roles. In this review, we discuss current understanding of the diverse functions of B7-H3 in carcinogenesis and cancer progression, and consider future directions for designing cancer immunotherapeutic agents targeting B7-H3.


Subject(s)
B7 Antigens/immunology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Neoplasms/immunology , B7 Antigens/biosynthesis , B7 Antigens/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Immunotherapy, Adoptive , Lymphocyte Activation/immunology , Neoplasms/therapy , Tumor Escape/immunology
13.
Int J Gynecol Cancer ; 24(3): 534-40, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24557437

ABSTRACT

OBJECTIVE: AT-rich interactive domain 1A (ARID1A) is a tumor suppressor gene that encodes the BAF250a protein. Recent studies have shown the loss of ARID1A expression in several types of tumors. We aimed to investigate the clinical and pathologic role of BAF250a in endometrial carcinoma. METHODS: We examined the expression of BAF250a and its correlation with the expression of p53, estrogen receptor, progesterone receptor, glucocorticoid receptor, hypoxiainduciblefactor-1α, and vascular endothelial growth factor in normal and various malignant endometrial tissues. RESULTS: The expression of BAF250 was significantly down-regulated in endometrial carcinoma when compared with normal endometrial tissues. The loss of BAF250a expression was found in 25% of endometrial carcinoma samples but not in normal endometrial tissues, complex endometrial hyperplasia, and atypical endometrial hyperplasia samples. Subtypes of endometrial carcinoma, especially uterine endometrioid carcinoma and uterine clear cell carcinoma, had higher frequency of loss of BAF250a expression. In addition, the expression of BAF250a was positively correlated with estrogen receptor and negatively correlated with p53 in poorly differentiated endometrial adenocarcinoma. Moreover, the expression of BAF250a was significantly associated with the differentiation status of endometrial carcinoma but not associated with clinical stage, the depth of myometrial invasion, lymph node metastasis, and overall survival of patients with endometrial carcinoma. CONCLUSIONS: Our data showed that loss of BAF250a is frequently found in high-grade endometrioid and clear cell carcinomas but not in other types of endometrial carcinoma. The loss of BAF250a expression does not have prognostic value for endometrial carcinoma.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma/pathology , Endometrial Neoplasms/pathology , Endometrium/pathology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Carcinoma/genetics , Carcinoma/metabolism , DNA-Binding Proteins , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrium/metabolism , Female , Humans , Middle Aged , Prognosis
14.
Cancer Immunol Immunother ; 60(7): 1047-55, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21519829

ABSTRACT

A retrospective cohort study including 112 patients suffering from esophageal squamous cell carcinoma (ESCC) was performed to investigate the expression of B7-H4 in ESCC and determine its association with patient's clinicopathological parameters and survival. Expression levels of B7-H4 on tumor cells and densities of tumor infiltrating lymphocytes (TILs) in the surgical specimens of ESCC tissues were characterized using immunohistochemical assays. Uni- and multivariate analyses were performed to evaluate the prognostic value of B7-H4 expression levels and densities of TILs in tumor sections. Positive B7-H4 immunostaining was observed in 107 of 112 (95.5%) of ESCC tissue sections. We further divided all patients into two major subgroups, a lower B7-H4 expression group with 46 patients and a higher B7-H4 expression group with 66 patients. We found that expression levels of B7-H4 on tumor cells were significantly correlated with patient's gender (P = 0.0288), distant metastasis (P = 0.0500), and TNM stage (P = 0.0258). Moreover, tumor cell B7-H4 expression was inversely correlated with densities of CD3(+) T cells in tumor nest (P = 0.0424) and CD8(+) T cells in tumor stroma (P = 0.0229). The overall survival rate of the patients with higher B7-H4 expression was significantly worse than that of the patients with lower B7-H4 expression (P = 0.0105, Hazard Ratio: 1.854, 95%CI:1.152-2.902). Markers of cell-mediated immune responses such as CD3, CD8, and T-bet were associated with better patient survival. The present study demonstrated that B7-H4 expression in human ESCC is associated with cancer progression, reduced tumor immunosurveillance and worse patient outcomes. B7-H4 can serve as a novel prognostic predictor for human ESCC and a potential target for the immune therapy against this malignancy.


Subject(s)
B7-1 Antigen/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Lymphocytes, Tumor-Infiltrating/pathology , Animals , Disease Progression , Female , Humans , Immunoenzyme Techniques , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Mice , Mice, Inbred BALB C , Middle Aged , Prognosis , Survival Rate , Tumor Microenvironment , V-Set Domain-Containing T-Cell Activation Inhibitor 1
15.
Metab Brain Dis ; 26(3): 185-94, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21826472

ABSTRACT

Apolipoprotein E (apoE) is one of the major transporters of cholesterol in the body and is essential for maintaining various neural functions in the brain. Given that hypercholesterolemia is a risk factor in Alzheimer's disease (AD), it has been suggested that altered cholesterol metabolism may be involved in the development of the pathogenesis, including neural degeneration, commonly observed in AD patients. Neurotrophic factors and their receptors, which are known to regulate various neural functions, are also known to be altered in various neurodegenerative diseases. We therefore hypothesized that cholesterol metabolism may itself influence the neurotrophin system within the brain. We decided to investigate this possibility by modulating the amount of dietary cholesterol given to apoE-knockout (apoE-KO) and wild-type (WT) mice, and examining the mRNA expression of various neurotrophin ligands and receptors in their hippocampal formations. Groups of eight-week-old apoE-KO and WT mice were fed a diet containing either "high" (HCD) or "normal" (ND) levels of cholesterol for a period of 12 weeks. We found that high dietary cholesterol intake elevated BDNF mRNA expression in both apoE-KO and WT mice and TrkB mRNA expression in apoE-KO animals. On the other hand, NGF and TrkA mRNA levels remained unchanged irrespective of both diet and mouse type. These findings indicate that altered cholesterol metabolism induced by HCD ingestion combined with apoE deficiency can elicit a differential response in the various neurotrophin ligand/receptor systems in the mouse hippocampus. Whether such changes can lead to neural degeneration, and the mechanisms that may be involved in this, awaits further research.


Subject(s)
Apolipoproteins E/deficiency , Brain-Derived Neurotrophic Factor , Cholesterol, Dietary , Hippocampus/metabolism , Receptor, trkB , Alzheimer Disease/metabolism , Animals , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cholesterol, Dietary/adverse effects , Cholesterol, Dietary/metabolism , Humans , Hypercholesterolemia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factor/drug effects , Nerve Growth Factor/metabolism , RNA, Messenger/metabolism , Receptor, trkA/drug effects , Receptor, trkA/metabolism , Receptor, trkB/drug effects , Receptor, trkB/metabolism
16.
Clin Chem Lab Med ; 48(6): 855-61, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20345231

ABSTRACT

BACKGROUND: Currently, no satisfactory biomarkers are available to screen for esophageal squamous cell carcinoma (ESCC). The goal of this study was to find biomarkers and establish a serum protein fingerprint model for early diagnosis of ESCC using the ClinProt protocol of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS). METHODS: Serum samples were collected from 62 patients with ESCC, nine patients with esophageal adenocarcinoma (EA) and 38 healthy individuals. Proteomic spectra of mass to charge ratio (m/z) were generated following the application of plasma to weak cationic-exchanger magnetic beads (WCX-MB). The spectral data were analyzed using a support vector machine, and potential biomarkers were chosen for system training and used to construct diagnostic models. RESULTS: Three differential patterns were established using MALDI-TOF MS. Pattern 1, consisting of 11 protein peaks, separated ESCC patients from the healthy individuals with a sensitivity of 90.0% and a specificity of 88.4%. Pattern 2, consisting of eight protein peaks, separated ESCC in stage I and stage II from stage III and stage IV with a sensitivity of 92.9% and a specificity of 82.3%. Pattern 3, consisting of seven protein peaks, separated ESCC from EA with a sensitivity of 91.3% and a specificity of 80.0%. CONCLUSIONS: These results suggested that MALDI-TOF MS combined with MB separation yields significantly higher sensitivity and specificity for the detection of serum protein in patients with ESCC.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Squamous Cell/diagnosis , Esophageal Neoplasms/diagnosis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Adenocarcinoma/blood , Adenocarcinoma/diagnosis , Adult , Aged , Carcinoma, Squamous Cell/blood , Esophageal Neoplasms/blood , Humans , Magnetics , Middle Aged , Neoplasm Staging , Proteome/analysis
17.
Biomed Pharmacother ; 121: 109611, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31731196

ABSTRACT

BACKGROUND: Our previous studies have showed that p-Hydroxylcinnamaldehyde (CMSP) could induce the differentiation of ESCC cells via the cAMP-RhoA-MAPK signalling pathway, which suggests a new potential strategy for ESCC treatment. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potent inducer of apoptosis in several tumour cells by binding to the death receptors DR4 and DR5. However, TRAIL has little effect on oesophageal squamous cell carcinoma (ESCC) cells due to the loss of the receptors. The present study determined the effect of CMSP, the firstly found chemical constituent of Cochinchinamomordica seed (CMS), on TRAIL-induced apoptosis and its mechanism in ESCC cells. METHODS: MTS assays were performed to examine the CMSP- and TRAIL-mediated inhibition of ESCC cell growth. Flow cytometry and Hoechst 33258 staining assays were used to detect apoptosis in ESCC cells treated with CMSP combined with TRAIL. Western blotting was used to determine the effect of CMSP on the expression of p38, p-p38, DR4, DR5, Bid and caspase-3/8 in ESCC cells treated with CMSP combined with TRAIL. Additionally, immunodeficient Balb-c/null mouse model was used to determine the chemotherapeutic efficacy of CMSP and TRAIL against ESCC tumour xenograft growth in vivo. RESULTS: We found that the combination of CMSP and TRAIL had a greater inhibitory effect on ESCC cell viability in vitro than CMSP or TRAIL alone. CMSP enhanced the TRAIL-induced apoptosis in ESCC cells by upregulating the expression of DR4 and DR5 via the p38 MAPK signalling pathway. Furthermore, the increased expression of DR4 and DR5 upon TRAIL-induced apoptosis in ESCC cells was mediated at least in part by subsequent caspase-3 and caspase-8 activation. Moreover, the in vivo model showed that tumour growth was significantly slower in CMSP and TRAIL combination-treated mice than in mice treated with CMSP or TRAIL alone. CONCLUSION: Taken together, our findings indicate that CMSP as an extract from TCM, might be as a potential sensitizer of TRAIL and thus provide a novel strategy for the clinical treatment of ESCC.


Subject(s)
Cinnamates/pharmacology , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Momordica/chemistry , TNF-Related Apoptosis-Inducing Ligand/pharmacology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Humans , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred BALB C , Receptors, TNF-Related Apoptosis-Inducing Ligand/analysis , Seeds/chemistry
18.
Cancer Invest ; 27(6): 604-12, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19440933

ABSTRACT

AIM: The Wnt signaling pathway plays a pivotal role in cellular developmental processes and human carcinogenesis. The aim of this study was to investigate the effects of quercetin on the growth of the colon carcinoma cell line and the regulation effect of quercetin on the Wnt/beta-catenin signaling pathway. METHODS: MTT assay was used to determine the reduction of cell viability of quercetin on SW480 cells and clone 26 cells. The apoptotic rate and cell-cycle analysis after treatment with quercetin was determined by flow cytometry. Effects of quercetin on mRNA expression of cyclin D(1) and survivin were detected by semiquantitative RT-PCR. After treatment with quercetin, the protein expression of cyclin D(1) and survivin in SW480 cells was analyzed by Western blot analysis. We built a Wnt/beta-catenin signaling pathway reporter gene model. The regulation effect of quercetin on the Wnt/beta-catenin signaling transcription was investigated by using this reporter gene model. RESULTS: Quercetin reduced cell viability in a dose- and time-dependent manner in SW480 and clone 26 cells. The percentages of SW480 cells and clone 26 cells at G(2)/M phase were increased significantly after treatment with 40 approximately 80 micromol/L quercetin for 48 hours. Quercetin induced the apoptosis of SW480 cells in a dose-dependent manner at the concentration of 20, 40, 60, anf 80 micromol/L. However, quercetin only induced the apoptosis of clone 26 cells at the concentration of 80 micromol/L. Quercetin downregulated transcriptional activity of beta-catenin/Tcf in SW480 cells transiently transfected with the TCF-4 reporter gene. Within 24 hours of treatment, a 160-mumol/L concentration of quercetin reduced beta-catenin/Tcf transcriptional activity by about 18-fold. Cyclin D(1) and the survivin gene were downregulated markedly by quercetin in a dose-dependent manner at both the transcription and protein expression levels. CONCLUSION: The results indicate that the molecular mechanism underlying the antitumor effect of quercetin in SW480 colon cancer cells is related to the inhibition of expression of cyclin D(1) and survivin as well as the Wnt/beta-catenin signaling pathway. Therefore, the Wnt/beta-catenin signaling pathway could be qualified as one of the promising targets for innovative treatment strategies of colorectal cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Cyclin D1/metabolism , Microtubule-Associated Proteins/metabolism , Quercetin/pharmacology , Signal Transduction/drug effects , Wnt Proteins/metabolism , beta Catenin/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Survival/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cyclin D1/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/genetics , RNA, Messenger/metabolism , Survivin , Transcription, Genetic/drug effects , Transfection
19.
Cancer Invest ; 27(1): 17-23, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19160097

ABSTRACT

Although non-steroidal anti-inflammatory drugs (NSAIDs) have been demonstrated to have cancer-preventive effects and induce apoptosis of cancer cells, the mechanism of their effects is not clearly known. We studied the mechanism in human esophageal cancer cell line TE13. The esophageal squamous cell carcinoma cell line TE-13 was cultured with NS-398 at different concentrations or for different times. Proliferation and apoptosis were measured by MTT reduction and flow cytometry. Prostaglandin F(1alpha) was determined with radioimmunoassay. Expression of COX-2 mRNA was measured by RT-PCR and COX-2 protein levels with Western blot analysis. Nuclear NF-kappaB and cytoplasmic IkappaB protein levels were determined by electrophoretic mobility shift assay and Western blot, respectively. NS-398 significantly inhibited cell proliferation and induced apoptosis at concentrations of 0.001, 0.01, 1, and 100 micromol/L. NS-398 dose-dependently decreased the levels of COX-2 mRNA, COX-2 protein, nuclear NF-kappaB protein and production of PGF(1alpha) and increased the cytoplasmic IkappaB protein. In conclusion, NS-398 inhibits the proliferation of, and induced apoptosis in, the cultured TE-13 SCC cell line. These changes correlate with a reduction in COX-2 mRNA and protein expression, prostaglandin synthesis, an inhibition of NF-kappaB nuclear translocation, and an increase in cytoplasmic IkappaB.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Cyclooxygenase 2/metabolism , Esophageal Neoplasms/pathology , NF-kappa B/antagonists & inhibitors , Nitrobenzenes/pharmacology , Sulfonamides/pharmacology , Blotting, Western , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/genetics , Cytoplasm/metabolism , Electrophoretic Mobility Shift Assay , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/metabolism , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
20.
Zhonghua Zhong Liu Za Zhi ; 31(7): 505-9, 2009 Jul.
Article in Zh | MEDLINE | ID: mdl-19950697

ABSTRACT

OBJECTIVE: To examine the in vivo anti-metastatic effect of enhanced expression of CD40L cDNA in murine ovarian cancer OVHM cells (CD40L-OVHM) injected into the spleen on liver metastasis in mice. METHODS: OVHM cells were inoculated into the spleen of 6 to 8 week-old female B6C3F1 (C57BL/6N x C3H/He) mice. The established liver metastasis was identified by histopathology (HE staining). OVHM cells, DNA-pMKITneo-OVHM cells or CD40L-OVHM cells were inoculated into the spleen of female B6C3F1 mice and the expressions of CD11c in splenic cells were detected by flow cytometry. The specific cytotoxicity of splenic cells was detected by MTT assay, and the serum cytokines of IFN-gamma, TNF-alpha, IL-12, IL-4 and IL-10 of the mice were measured by enzyme linked immunoabsorbent assay. The liver metastases and the survival time of the mice were also recorded. RESULTS: The mouse models with liver metastasis by injecting tumor cells into the spleen of mice were established. The expression of CD11c and the specific killing rate in CD40L-OVHM cells group was significantly higher than that in the OVHM cells group and DNA-pMKITneo-OVHM cells group. The expressions of IFN-gamma, TNF-alpha and IL-12 in the CD40L-OVHM cells group were much more increased than OVHM cells group and DNA-pMKITneo-OVHM cells group, but the expressions of IL-4 and IL-10 in the CD40L-OVHM cells group were decreased significantly (p < 0.05). The average weights of livers and spleens of mice in CD40L-OVHM cells group were significantly lower than those of DNA-pMKITneo-OVHM cells group and OVHM cells group. The survival time of mice in CD40L-OVHM cells group was also significantly longer than that in the OVHM cells group and DNA-pMKITneo-OVHM cells group (P < 0.05). CONCLUSION: The data directly demonstrate that the expression of CD40L in ovarian cancer cells (CD40L-OVHM) can enhance the proliferation and differentiation of dendritic cells in the spleen and induce specific cytotoxic effect of T cells in the spleen, and may regulate the immune function of peripheral blood cells and the immune balance between Th1 cells and Th2 cells, which maybe the possible mechanism induced by CD40L in mice inhibiting the development of liver metastasis.


Subject(s)
CD11c Antigen/metabolism , CD40 Ligand/metabolism , Liver Neoplasms/secondary , Ovarian Neoplasms/metabolism , Animals , CD40 Ligand/genetics , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , DNA, Complementary/genetics , Dendritic Cells/cytology , Female , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-12/metabolism , Interleukin-4/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Spleen/metabolism , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL