Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 71(2): 201-215.e7, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30029001

ABSTRACT

Macrophages are a dominant leukocyte population in the tumor microenvironment and actively promote cancer progression. However, the molecular mechanism underlying the role of macrophages remains poorly understood. Here we show that polarized M2 macrophages enhance 3-phosphoinositide-dependent protein kinase 1 (PDPK1)-mediated phosphoglycerate kinase 1 (PGK1) threonine (T) 243 phosphorylation in tumor cells by secreting interleukin-6 (IL-6). This phosphorylation facilitates a PGK1-catalyzed reaction toward glycolysis by altering substrate affinity. Inhibition of PGK1 T243 phosphorylation or PDPK1 in tumor cells or neutralization of macrophage-derived IL-6 abrogates macrophage-promoted glycolysis, proliferation, and tumorigenesis. In addition, PGK1 T243 phosphorylation correlates with PDPK1 activation, IL-6 expression, and macrophage infiltration in human glioblastoma multiforme (GBM). Moreover, PGK1 T243 phosphorylation also correlates with malignance and prognosis of human GBM. Our findings demonstrate a novel mechanism of macrophage-promoted tumor growth by regulating tumor cell metabolism, implicating the therapeutic potential to disrupt the connection between macrophages and tumor cells by inhibiting PGK1 phosphorylation.


Subject(s)
Macrophages/metabolism , Phosphoglycerate Kinase/genetics , Phosphoglycerate Kinase/metabolism , 3-Phosphoinositide-Dependent Protein Kinases/genetics , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Glycolysis , Humans , Macrophages/pathology , Mice , Mice, Nude , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Phosphorylation , Prognosis , Tumor Microenvironment
2.
Gut ; 70(5): 951-961, 2021 05.
Article in English | MEDLINE | ID: mdl-32998878

ABSTRACT

OBJECTIVE: Tumour pathology contains rich information, including tissue structure and cell morphology, that reflects disease progression and patient survival. However, phenotypic information is subtle and complex, making the discovery of prognostic indicators from pathological images challenging. DESIGN: An interpretable, weakly supervised deep learning framework incorporating prior knowledge was proposed to analyse hepatocellular carcinoma (HCC) and explore new prognostic phenotypes on pathological whole-slide images (WSIs) from the Zhongshan cohort of 1125 HCC patients (2451 WSIs) and TCGA cohort of 320 HCC patients (320 WSIs). A 'tumour risk score (TRS)' was established to evaluate patient outcomes, and then risk activation mapping (RAM) was applied to visualise the pathological phenotypes of TRS. The multi-omics data of The Cancer Genome Atlas(TCGA) HCC were used to assess the potential pathogenesis underlying TRS. RESULTS: Survival analysis revealed that TRS was an independent prognosticator in both the Zhongshan cohort (p<0.0001) and TCGA cohort (p=0.0003). The predictive ability of TRS was superior to and independent of clinical staging systems, and TRS could evenly stratify patients into up to five groups with significantly different prognoses. Notably, sinusoidal capillarisation, prominent nucleoli and karyotheca, the nucleus/cytoplasm ratio and infiltrating inflammatory cells were identified as the main underlying features of TRS. The multi-omics data of TCGA HCC hint at the relevance of TRS to tumour immune infiltration and genetic alterations such as the FAT3 and RYR2 mutations. CONCLUSION: Our deep learning framework is an effective and labour-saving method for decoding pathological images, providing a valuable means for HCC risk stratification and precise patient treatment.


Subject(s)
Carcinoma, Hepatocellular/pathology , Deep Learning , Liver Neoplasms/pathology , Prognosis , Aged , Carcinoma, Hepatocellular/mortality , Female , Humans , Liver Neoplasms/mortality , Male , Middle Aged , Neoplasm Staging , Phenotype , Survival Analysis
3.
Oncologist ; 26(4): e524-e529, 2021 04.
Article in English | MEDLINE | ID: mdl-32997436

ABSTRACT

This article reports a case of advanced metastatic low-grade sarcoma. The patient was diagnosed with an inoperable large (14 × 12 cm) lesion on his neck in September 2015 and underwent two ineffective chemotherapies in the following 4 months. Interestingly, although several pathologists could not agree on the histopathological diagnosis, the precise molecular pathological diagnosis was obtained using next-generation sequencing (NGS) and finally brought excellent therapeutic effects. The patient was detected to have CARS-ALK fusion by NGS and then was successfully treated with crizotinib orally. He received surgical resection of primary and metastatic lesions after tumor shrinkage. The combined treatment brought a durable response for 40 months. Although the tumor recurred in July 2019, the patient has been responding well to the second-line ALK tyrosine kinase inhibitor alectinib to date. We performed whole genome sequencing on the patient's primary, metastatic, and recurrent tumors and did comprehensive genomic analysis. Furthermore, our analysis results revealed that a whole genome duplication event might have happened during tumorigenesis of this case. KEY POINTS: To our best knowledge, this is the first report of a very successful treatment with first- and second-line ALK tyrosine kinase inhibitors for CARS-ALK fusion-positive metastatic low-grade sarcoma. Molecular pathological result can guide precision treatment for sarcoma, even when the exact histopathology cannot be obtained. Multiple samples from this patient were analyzed using whole genome sequencing. Results provided detailed genomic characteristics and showed tumor evolution of this low-grade sarcoma case. A whole genome duplication event might have happened during tumorigenesis of this low-grade sarcoma case.


Subject(s)
Lung Neoplasms , Sarcoma , Anaplastic Lymphoma Kinase/genetics , Genomics , Humans , Male , Neoplasm Recurrence, Local , Protein Kinase Inhibitors/therapeutic use
4.
PLoS Biol ; 16(10): e2006483, 2018 10.
Article in English | MEDLINE | ID: mdl-30335765

ABSTRACT

Glycogen metabolism commonly altered in cancer is just beginning to be understood. Phosphoglucomutase 1 (PGM1), the first enzyme in glycogenesis that catalyzes the reversible conversion between glucose 1-phosphate (G-1-P) and glucose 6-phosphate (G-6-P), participates in both the breakdown and synthesis of glycogen. Here, we show that PGM1 is down-regulated in hepatocellular carcinoma (HCC), which is associated with the malignancy and poor prognosis of HCC. Decreased PGM1 expression obstructed glycogenesis pathway, which leads to the increased flow of glucose into glycolysis, thereby promoting tumor cell proliferation and HCC development. The loss of forkhead box protein J2 (FOXJ2), at least partly due to low genomic copy number in HCC, releases cellular nucleic acid-binding protein (CNBP), a nucleic acid chaperon, to bind to and promote G-quadruplex formation in PGM1 promoter and therefore decreases PGM1 expression. In addition, integrated analyses of PGM1 and FOXJ2 expression provide a better prediction for the malignance and prognosis of HCC. This study establishes a tumor-suppressive role of PGM1 by regulating glucose trafficking and uncovers a novel regulatory mechanism of PGM1 expression.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Glucose/metabolism , Liver Neoplasms/metabolism , Phosphoglucomutase/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Disease Progression , Down-Regulation , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glycolysis , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Middle Aged , Phosphoglucomutase/deficiency , Phosphoglucomutase/genetics , Prognosis , Promoter Regions, Genetic , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
5.
Zhonghua Nan Ke Xue ; 27(12): 1064-1068, 2021 Dec.
Article in Zh | MEDLINE | ID: mdl-37454313

ABSTRACT

Objective: To investigate the protective effect of long-term consumption of hydrogen-rich water (HRW) on the percentage of progressively motile sperm (PMS) in male rats. METHODS: Twenty normal healthy male SD rats were equally randomized into an HRW and a control group, the former given HRW (1.2 ppm) and the latter normal saline, both intragastrically at 2 ml/d for 9 months. Then, the bilateral epididymides of the rats were harvested for preparation of sperm suspension and detection of the percentage of PMS. The testis tissue was isolated for HE staining and determination of the expressions of the Ki67, CYBB, eNOS, CLDN3 and SRD5A2 proteins using the streptavidin-peroxidase (SP) immunohistochemical method. RESULTS: The percentage of PMS was significantly higher in the HRW than in the control group (ï¼»64.3 ± 4.7ï¼½% vs ï¼»55.3 ± 9.5ï¼½%, P < 0.05), and so was the expression of Ki67 in the testicular tissue (P < 0.01). Compared with the controls, the rats in the HRW group showed markedly decreased oxidative stress-related index CYBB (P < 0.01), increased eNOS level (P < 0.01), and upregulated expressions of sperm development-related proteins CLDN3 and SRD5A2 (P < 0.01 and P < 0.05). CONCLUSIONS: Hydrogen not only regulates the expressions of some oxidative stress-related indicators, but also increases the expressions of the molecules promoting sperm maturation and motility, which provides a theoretical basis and experimental support for the application and studies of hydrogen in asthenospermia.

6.
EMBO J ; 35(6): 668-84, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26912724

ABSTRACT

A key step of Wnt signaling activation is the recruitment of ß-catenin to the Wnt target-gene promoter in the nucleus, but its mechanisms are largely unknown. Here, we identified FoxM1 as a novel target of Wnt signaling, which is essential for ß-catenin/TCF4 transactivation. GSK3 phosphorylates FoxM1 on serine 474 which induces FoxM1 ubiquitination mediated by FBXW7. Wnt signaling activation inhibits FoxM1 phosphorylation by GSK3-Axin complex and leads to interaction between FoxM1 and deubiquitinating enzyme USP5, thereby deubiquitination and stabilization of FoxM1. FoxM1 accumulation in the nucleus promotes recruitment of ß-catenin to Wnt target-gene promoter and activates the Wnt signaling pathway by protecting the ß-catenin/TCF4 complex from ICAT inhibition. Subsequently, the USP5-FoxM1 axis abolishes the inhibitory effect of ICAT and is required for Wnt-mediated tumor cell proliferation. Therefore, Wnt-induced deubiquitination of FoxM1 represents a novel and critical mechanism for controlling canonical Wnt signaling and cell proliferation.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Wnt Proteins/metabolism , beta Catenin/metabolism , Cell Line , Endopeptidases/metabolism , Forkhead Box Protein M1 , Humans , Transcriptional Activation , Ubiquitination , Wnt Signaling Pathway
7.
World J Surg Oncol ; 18(1): 318, 2020 Dec 04.
Article in English | MEDLINE | ID: mdl-33276775

ABSTRACT

BACKGROUND: The proline-rich protein 11 (PRR11) is a newly identified oncogene associated with a poor prognosis in several human cancers. Nonetheless, research on its role in ovarian cancer (OC) remains largely understudied. Therefore, this study aims to evaluate the expression levels of PRR11 protein and its role in human ovarian cancer. METHODS: Immunohistochemistry analysis was used to evaluate the expression levels of PRR11 protein in human samples obtained from 49 patients diagnosed with OC and subjected to curative surgery in the First Affiliated Hospital of Wenzhou Medical University between 2007 and 2015. RESULTS: In total, 57.1% of the primary OC tumor tissue evaluated demonstrated overexpression of PRR11. Meanwhile, the survival analysis showed that the overall survival (OS) of patients presenting overexpression of PRR11 was significantly lower than the OS of the patients with negative PRR11. In subsequent experiments, it was found that silencing the expression of PRR11 expression inhibited the proliferation of tumor cells and the migration of cells in vitro. Further, cells subjected to PRR11 knockdown exhibited a decrease in tumor growth in vivo. The downregulation of PRR11 was coupled with a decrease in N-cadherin and downregulation in the expression of early growth response protein 1 (EGR1). CONCLUSIONS: The findings suggest that PRR11 might be considered as a potential target for prognostic assessment and gene therapy strategies for patients diagnosed with OC.


Subject(s)
Ovarian Neoplasms , Carcinoma, Ovarian Epithelial , Female , Humans , Phenotype , Prognosis , Proline
8.
Cancer Cell Int ; 19: 74, 2019.
Article in English | MEDLINE | ID: mdl-30976198

ABSTRACT

BACKGROUND: Gastric cardia adenocarcinoma (GCA) is an aggressive subtype of gastric cancer with a high metastatic rate. However, the metastatic biomarker of GCA has not been established. METHODS: To search for the biomarker for GCA metastasis, we here examined expression of the Hippo signaling effector WWTR1 (WW domain containing transcription regulator 1, commonly listed as TAZ) in tumor tissue samples from 214 GCA cases using the tissue microarray assay (TMA), and statistically analyzed association of the WWTR1 expression with metastasis-related pathological outcomes and cumulative survival of the GCA patients. Furthermore, shRNA knockdown was used to determine the role of WWTR1 in promoting cell migration in gastric cancer cells. RESULTS: The results have shown that WWTR1 is overexpressed in 66.4% of the GCA tumor samples. Expression of WWTR1 has a significant inverse correlation with cumulative survival of GCA patients (p < 0.01). WWTR1 positive patients had a mean survival of 56.9 ± 4.4 months, comparing to WWTR1 negative mean survival of 77.3 ± 5.9 months. More importantly, expression of WWTR1 significantly associated with tumor invasion and metastasis (in T stage, p = 0.031; N stage, p < 0.01; and TNM stage, p < 0.001). Furthermore, knockdown of WWTR1 impaired migration of gastric cancer AGS cells. CONCLUSIONS: Our studies have identified WWTR1 as a metastatic biomarker of GCA for poor prognosis, defined a role of WWTR1 in driving metastasis of gastric cancer, and suggested WWTR1 as a potential target for anti-metastatic therapy of GCA.

9.
BMC Cancer ; 18(1): 206, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29463236

ABSTRACT

BACKGROUND: Gastric cancer patients with widespread metastasis, especially meningeal metastases, have an extremely prognosis and limited therapeutic choices. CASE PRESENTATION: We reported the case of a 39-year-old male patient with HER2-positive gastric cancer with bone and meningeal metastases. He presented with multiple bone metastases and received 3 cycles of docetaxel plus S1. However, he complained with headache and imaging examinations revealed leptomeningeal carcinomatosis. FISH revealed that tumor cells in the cerebrospinal fluid were HER-positive. Herceptin was added to the regimen, but the symptoms were not relieved, the patient suffered from dizziness and nausea. The chemotherapy regimen was switched d to lapatinib (orally at 1250 mg/day, every day), capecitabine (orally at 1000 mg/m2, bid for 2 weeks, followed by a 1-week rest interval, as 1 cycle) and Herceptin (390 mg/3 weeks). After 3 weeks of the new treatment, all the symptoms relieved. The clinical complete response was maintained for 3 months. CONCLUSIONS: Lapatinib/Capecitabine combination therapy is an alternative treatment strategy for leptomeningeal carcinomatosis of HER2-positive gastric cancer in which trastuzumab and/or chemotherapy essentially has no effect.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Meningeal Carcinomatosis/drug therapy , Meningeal Carcinomatosis/secondary , Stomach Neoplasms/pathology , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers , Biomarkers, Tumor , Capecitabine/administration & dosage , Diagnostic Imaging , Humans , In Situ Hybridization, Fluorescence , Lapatinib , Male , Meningeal Carcinomatosis/diagnosis , Multimodal Imaging , Neoplasm Metastasis , Quinazolines/administration & dosage , Receptor, ErbB-2/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Trastuzumab/administration & dosage , Treatment Outcome
10.
BMC Cancer ; 18(1): 570, 2018 May 18.
Article in English | MEDLINE | ID: mdl-29776401

ABSTRACT

BACKGROUND: A-disintegrin and metalloproteinases (ADAMs) are members of a family of multidomain transmembrane and secreted proteins. Specific ADAMs are upregulated in human cancers and correlated with tumor progression and poor outcome, but rarely studied in human hilar cholangiocarcinoma (HC). This study aimed to explore the expression profiles of ADAMs and their potential underlying mechanisms promoting cancer progression. METHODS: mRNA expression of ADAM-9, - 10, - 11, - 12, - 15, - 17, - 28, and - 33 was analyzed in human hilar cholangiocarcinoma (HC) samples. Immunohistochemical (IHC) analysis was used to detect the expression of ADAM-10, - 17, - 28, and FoxM1 in HC. The regulation of ADAM-17 by FoxM1 and their functional study was investigated in vivo and in vitro. RESULTS: ADAM-10, - 17, and - 28 were upregulated in tumors compared with matched non-cancerous tissues. IHC analysis revealed increased expression of ADAM-10, - 17, and - 28 in HC cells, and ADAM17 seems to be an independent prognostic factor. ADAM-17 is regulated by FoxM1. A decrease in the expression of ADAM-17 by silencing FoxM1 led to an inhibition of cell proliferation, tumor growth, and the production of tumor necrosis factor α. IHC analysis showed co-expression of FoxM1 and ADAM-17 in HC specimens. CONCLUSIONS: The findings of the present study show an important role of the cross-talk among FoxM1, ADAM-17, and TNFa in HC development and progression.


Subject(s)
ADAM17 Protein/metabolism , Bile Duct Neoplasms/genetics , Biomarkers, Tumor/genetics , Forkhead Box Protein M1/metabolism , Klatskin Tumor/genetics , ADAM17 Protein/genetics , Bile Duct Neoplasms/mortality , Bile Duct Neoplasms/pathology , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Disease Progression , Female , Forkhead Box Protein M1/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Klatskin Tumor/mortality , Klatskin Tumor/pathology , Male , Prognosis , RNA, Messenger/metabolism , Survival Analysis , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
11.
J Surg Res ; 220: 223-233, 2017 12.
Article in English | MEDLINE | ID: mdl-29180185

ABSTRACT

BACKGROUND: The ADAMs proteases are a multifunctional family of proteins, many of which participate in the pathogenesis of cancers. The expression and regulation of ADAMs has not yet been fully examined in gastric cancer. MATERIALS AND METHODS: Using reverse transcription-PCR, messenger RNA expression of ADAM-9, ADAM-10, ADAM-11, ADAM-12, ADAM-15, ADAM-17, ADAM-28, and ADAM-33 was detected in gastric cancer. ADAM-10, ADAM-17, ADAM-28, and FoxM1 expression in gastric cancer was detected by Western blot and immunohistochemistry. The correlation between ADAM-17 and FoxM1 was analyzed in vivo and in vitro. RESULTS: The messenger RNA levels of ADAM-9, ADAM-10, ADAM-15, ADAM-17, ADAM-28, and ADAM-33 were increased in tumor tissues compared with adjacent normal tissue, especially ADAM-10, ADAM-17, and ADAM-28. FoxM1 expression correlated significantly with ADAM-17 expression. FoxM1 regulates ADAM-17 expression in vivo and in vitro, which in turn influences proliferation and tumor growth of gastric cancer cells. Furthermore, Cox regression analysis revealed that FoxM1 and ADAM-17 are independent prognostic factors for patients with gastric cancer. CONCLUSIONS: These results indicate that overexpression of ADAMs may contribute to gastric cancer progression and that ADAM-17 is a downstream target of FoxM1. Overall, the present study highlights the potential for FoxM1 and ADAMs as potential therapeutic targets for patients with gastric carcinoma.


Subject(s)
ADAM17 Protein/metabolism , Adenocarcinoma/enzymology , Forkhead Box Protein M1/metabolism , Stomach Neoplasms/enzymology , Adenocarcinoma/mortality , Biomarkers, Tumor/metabolism , Cell Line, Tumor , China/epidemiology , Female , Humans , Male , Middle Aged , Stomach Neoplasms/mortality
12.
Cell Physiol Biochem ; 39(2): 768-79, 2016.
Article in English | MEDLINE | ID: mdl-27467187

ABSTRACT

BACKGROUND/AIMS: Neural precursor cell-expressed developmentally down-regulated gene 4 (NEDD4) plays an important role in tumor cell growth, yet its role in hepatocellular carcinoma (HCC) remains unclear. This study is to establish NEDD4 as a prognostic biomarker by which the survival of HCC patients can be predicted and to reveal the role of NEDD4 in hepatocellular carcinoma cell growth. METHODS: The expression of NEDD4 in 219 HCC specimens was assessed by immunohistochemistry. Postoperative overall survival and time to recurrence were evaluated by univariate and multivariate analyses. The roles of NEDD4 in hepatocellular carcinoma cell proliferation and invasion were determined. RESULTS: The patients with low NEDD4 expression tumors had an average cumulative survival of 64.9 ± 6.5 months during follow-up while the patients with high NEDD4 expression tumors had an average cumulative survival of 20.3 ± 15.8 months. NEDD4 silencing inhibited Huh7 cell proliferation and altered cell cytoskeletal assembly, and NEDD4 depletion furthermore seemed to suppress cell migration and invasion. A possible molecular mechanism for the observed effects might be that NEDD4 silence led to an increase in PTEN (phosphatase and tensin homologue) expression, which in turn resulted in the inactivation of STAT3, AKT, and ERK1/2. CONCLUSION: Our findings indicate that NEDD4 may participate in the HCC progression and may therefore be a potential target for HCC therapy.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Endosomal Sorting Complexes Required for Transport/metabolism , Liver Neoplasms/metabolism , PTEN Phosphohydrolase/metabolism , Ubiquitin-Protein Ligases/metabolism , Actin Cytoskeleton/metabolism , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/surgery , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Movement , Endosomal Sorting Complexes Required for Transport/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Liver Neoplasms/genetics , Liver Neoplasms/surgery , Male , Microscopy, Confocal , Middle Aged , Nedd4 Ubiquitin Protein Ligases , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Ubiquitin-Protein Ligases/genetics
13.
Tumour Biol ; 37(11): 14961-14967, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27651160

ABSTRACT

Homeobox C8 (HOXC8) has been implicated in cell growth, migration, and metastasis of various cancers, yet its role in osteosarcoma remains to be explored. In the present study, resected osteosarcoma specimens from 50 patients were enrolled to evaluate the expression of HOXC8 protein by immunohistochemistry (IHC). In vitro and in vivo assays were used to determine the effect of HOXC8 on cell growth, migration, and tumor growth. HOXC8 expression was observed in 31 (62.0 %) of the 50 primary tumors and significantly associated with poorly or un-differentiated specimens (P = 0.031) and larger tumor size (P = 0.049). Survival analysis demonstrated that HOXC8 is a candidate predictive factor in predicting patients' outcome and chemotherapeutic effect. HOXC8 knockdown led to inhibition of tumor cell proliferation and migration in vitro by inhibiting MMP-9 expression and tumor growth in vivo. Our results strongly suggest that HOXC8 is involved in the tumorigenesis of osteosarcoma and might serve as a novel predictor for patients' outcome.


Subject(s)
Bone Neoplasms/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Homeodomain Proteins/genetics , Matrix Metalloproteinase 9/biosynthesis , Osteosarcoma/pathology , Adolescent , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , RNA Interference , RNA, Small Interfering , Transplantation, Heterologous , Treatment Outcome
14.
Tumour Biol ; 37(4): 4803-11, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26520441

ABSTRACT

Activation of AMP-activated protein kinase (AMPK) suppressed mammalian target of rapamycin (mTOR) pathway, resulting in impaired cancer cell proliferation. Two cohorts (50 and 1072 cases) of patients with resected gastric adenocarcinoma were enrolled in the study. Immunohistochemical staining for p-AMPKa, p-ACC, p-mTOR, p-S6, and p-4EBP1 was performed on the 50-patient cohort. Tissue microarray blocks containing samples from 1072 patients of Chinese ethnicity were used for the immunohistochemical detection of p-AMPKa and p-S6 levels. p-AMPK and p-ACC were frequently inactivated in both cohorts of gastric cancer samples, while p-mTOR, p-S6, and p-4EBP1 were frequently activated in the small cohort of gastric cancer. However, only levels of p-AMPKa and p-S6 were associated with the overall survival of gastric cancer patients. In the larger 1072-patient cohort, downregulation of p-AMPKa and upregulation of p-S6 were associated with tumor progression and were independent predictors of survival after resection of primary gastric cancer. Therefore, reciprocal expression of p-AMPKa and p-S6 may be promising prognostic biomarkers in patients with gastric cancer.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Biomarkers, Tumor/biosynthesis , Ribosomal Protein S6 Kinases, 70-kDa/biosynthesis , Stomach Neoplasms/genetics , AMP-Activated Protein Kinases/genetics , Acetyl-CoA Carboxylase/biosynthesis , Acetyl-CoA Carboxylase/genetics , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Adult , Aged , Biomarkers, Tumor/genetics , Cell Cycle Proteins , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Neoplasm Proteins/biosynthesis , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Phosphorylation , Prognosis , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Signal Transduction , Stomach Neoplasms/pathology , TOR Serine-Threonine Kinases/biosynthesis
15.
Tumour Biol ; 37(3): 4159-67, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26490986

ABSTRACT

Secreted protein acidic and rich in cysteines-like protein 1 (SPARCL1) has been implicated in tumor initiation, formation, and progression of various cancers, yet its role in hilar cholangiocarcinoma remains largely uncharacterized. In the present study, tissue microarrays containing resected hilar cholangiocarcinoma specimens from 92 patients were used to evaluate the expression of SPARCL1 protein by immunohistochemistry (IHC). In vitro assays were used to determine the effect of SPARCL1 overexpression on cell growth and migration. Loss of SPARCL1 expression was observed in 46 (50.0 %) of the 92 primary tumors. SPARCL1 expression is inversely associated with poorly or undifferentiation specimens (P = 0.030) in addition to lymph node metastasis (P = 0.047). Survival analysis demonstrated that SPARCL1 is an independent factor in predicting the outcome of patients with hilar cholangiocarcinoma. SPARCL1 overexpression suppressed tumor cell migration in vitro by inhibiting MMP-9, MMP-2, Vimentin, and Fibronectin expression, whereas did not inhibit cell proliferation in vitro. Our results suggest that loss of SPARCL1 is involved in the tumorigenesis of hilar cholangiocarcinoma and may serve as a novel molecular biomarker for patients' outcome.


Subject(s)
Bile Duct Neoplasms/metabolism , Biomarkers, Tumor/metabolism , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Klatskin Tumor/metabolism , Neoplasm Recurrence, Local/metabolism , Adult , Aged , Bile Duct Neoplasms/mortality , Bile Duct Neoplasms/pathology , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Movement , Female , Humans , Kaplan-Meier Estimate , Klatskin Tumor/mortality , Klatskin Tumor/pathology , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local/mortality , Proportional Hazards Models
16.
Tumour Biol ; 37(11): 15079-15085, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27658779

ABSTRACT

The aim of this study was to investigate the prognostic value of dicarbonyl/L-xylulose reductase (DCXR) in human hepatocellular carcinoma (HCC). Immunohistochemistry and tissue microarrays were used to evaluate DCXR protein expression levels. Image-Pro Plus was used to calculate the integral optic density (IOD) in each tissue sample, which represented the expression level of DCXR. DCXR proteins were found to be significantly lower in HCC tumor tissues (P < 0.0001) according to immunohistochemical analysis of DCXR protein levels in 74 paired HCC tissue and peritumoral non-cancer tissues. The prognostic value of DCXR in HCC was assessed in 290 cases of the training cohort and 74 cases of the validation cohort. Shorter overall survival (OS) time and shorter time to recurrence (TTR) in both the training and validation set were found to be associated with lower expression levels of DCXR. In the training set, the expression level of DCXR in HCC was an independent prognostic factor for OS according to univariate and multivariate analyses. In conclusion, DCXR expression is an independent prognostic factor for OS and TTR of post-operative HCC patients, and low expression levels of DCXR in HCC may indicate poor outcome of HCC patients after surgical resection.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Sugar Alcohol Dehydrogenases/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/therapy , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Male , Middle Aged , Neoplasm Staging , Prognosis , Survival Rate , Tissue Array Analysis
17.
Mol Cancer ; 14: 193, 2015 Nov 14.
Article in English | MEDLINE | ID: mdl-26576639

ABSTRACT

BACKGROUND: The therapeutic and prognostic value of the glycolytic enzymes hexokinase, phosphofructokinase, and pyruvate kinase (PK) has been implicated in a variety of cancers, while their roles in treatment of and prognosis for hilar cholangiocarcinoma (HC) remain unclear. In this study, we determined the expression of PKM2 in and its impact on biology and clinical outcome of human HC. METHODS: The regulation and function of PKM2 in HC pathogenesis was evaluated using human tissues, molecular and cell biology, and animal models, and its prognostic significance was determined according to its impact on patient survival. RESULTS: We found that expression of hexokinase 1 and the M2 splice isoform of PK (PKM2) was upregulated in HC tissues and that this expression correlated with tumor recurrence and outcome. PKM2 expression was increased in HC cases with chronic cholangitis as demonstrated by isobaric tags for relative and absolute quantification. High PKM2 expression was highly correlated with high syndecan 2 (SDC2) expression and neural invasion. PKM2 downregulation led to a decrease in SDC2 expression. Treatment with metformin markedly suppressed PKM2 and SDC2 expression at both the transcriptional and posttranscriptional levels and inhibited HC cell proliferation and tumor growth. CONCLUSIONS: PKM2 regulates neural invasion of HC cells at least in part via regulation of SDC2. Inhibition of PKM2 and SDC2 expression contributes to the therapeutic effect of metformin on HC. Therefore, PKM2 is an independent prognostic factor and potential therapeutic target for human HC.


Subject(s)
Carrier Proteins/metabolism , Klatskin Tumor/metabolism , Klatskin Tumor/pathology , Membrane Proteins/metabolism , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Protein Isoforms/metabolism , Thyroid Hormones/metabolism , Adult , Aged , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Hexokinase/genetics , Hexokinase/metabolism , Humans , Klatskin Tumor/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Middle Aged , Neoplasm Recurrence, Local/genetics , Prognosis , Protein Isoforms/genetics , Syndecan-2/genetics , Syndecan-2/metabolism , Thyroid Hormones/genetics , Thyroid Hormone-Binding Proteins
18.
Mol Cancer ; 13: 248, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25395181

ABSTRACT

BACKGROUND: Gastric cardia adenocarcinoma (GCA) is the most aggressive subtype of gastric carcinoma. New molecular markers and therapeutic targets are needed for diagnosis, prognosis and treatment of GCA. This study is to establish the E3 ubiquitin ligase Nedd4-1 as a prognostic biomarker to predict the survival and guide the treatment of GCA patients. METHODS: Expression of Nedd4-1 in 214 GCA tumor samples was detected by immunohistochemistry staining (IHC) using tissue microarray assay (TMA). Association of Nedd4-1 with cumulative survival of the TNM stages I-III patients and clinicopathological characteristics was statistically analyzed. The role of Nedd4-1 in gastric cancer cell migration and invasion were determined by transwell and wound healing assays. RESULTS: Nedd4-1 is overexpressed in 83% of the GCA tumors. The 5-year survival rate in Nedd4-1 negative GCA patients is as high as 96%. Log-rank analysis indicated that overexpression of Nedd4-1 is inversely correlated with cumulative survival (χ(2) = 21.885, p <0.001). Multivariate logistic regression analysis showed that overexpression of Nedd4-1 is associated with an extremely low GCA survival rate with a hazard ratio (HR) = 0.068 (p = 0.008) in TNM stages I-III patients. Statistical analysis of association of Nedd4-1 overexpression with clinicopathological characteristics revealed that overexpression of Nedd4-1 is tightly associated with TNM stage (p < 0.001). Knockdown of Nedd4-1 in gastric cancer cell lines AGS and N87 dramatically inhibited the gastric cancer cell migration and invasion. CONCLUSIONS: Our results indicate that Nedd4-1 is an exceptional prognostic biomarker for GCA and suggest that Nedd4-1 may play an essential role in GCA metastasis.


Subject(s)
Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Neoplasm Metastasis/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics , Adenocarcinoma/pathology , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Female , HEK293 Cells , Humans , Male , Middle Aged , Nedd4 Ubiquitin Protein Ligases , Neoplasm Metastasis/pathology , Neoplasm Staging/methods , Prognosis , Stomach Neoplasms/pathology , Survival Rate
19.
BMC Cancer ; 14: 520, 2014 Jul 19.
Article in English | MEDLINE | ID: mdl-25038797

ABSTRACT

BACKGROUND: Annexin-1 contributes to the pathological consequence and sequelae of most serious human diseases including cardiovascular disease and cancer. Although diverse roles in carcinogenesis have been postulated, its role in human gastrointestinal cancers still remains controversial. METHODS: The mRNA and protein expression profiles of ANXA1 were studied in human esophageal, gastric, pancreatic, colorectal, liver, and bile duct cancers using Real-Time PCR, western blotting, and immunohistochemistry. Gain/loss-of-function by pcDNA3.1-ANXA1 and ANXA1-shRNA was performed in gastric cancer cells. RESULTS: ANXA1 was widely expressed in adult gastrointestinal tissue. All methods showed that ANXA1 was down-regulated in esophageal, gastric, and bile duct cancers, but up-regulated in pancreatic cancer. Forced ANXA1 expression in gastric cancer cells leads to cell growth inhibition and concomitantly modulates COX-2 expression. We confirm loss of ANXA1 and overexpression of COX-2 in clinical gastric cancer, suggesting that the anti-proliferative function of ANXA1 against COX-2 production might be lost. CONCLUSIONS: ANXA1 expression is "tumor-specific" and might play a multifaceted role in cancer development and progression. ANXA1 was widely expressed in normal gastrointestinal epithelium, suggesting its role in the maintenance of cellular boundaries. Furthermore, ANXA1 regulates GC cell viability via the COX-2 pathway.


Subject(s)
Annexin A1/genetics , Annexin A1/metabolism , Gastrointestinal Neoplasms/pathology , Gastrointestinal Tract/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclooxygenase 2/metabolism , Female , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Tract/pathology , Gene Expression Regulation, Neoplastic , Humans , Male
20.
iScience ; 27(2): 109011, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38357662

ABSTRACT

Loss of Protocadherin 9 (PCDH9) is associated with the metastasis and the prognosis of gastric cancer patients, while the molecular mechanism of PCDH9-impaired gastric cancer metastasis remains unclear. Here we show that PCDH9 is cleaved in gastric cancer cells. Intracellular domain of PCDH9 translocates into nucleus, where it interacts with DNA methyltransferase 1 (DNMT1) and increases DNMT1 activity. Activated DNMT1 downregulates cadherin 2 (CDH2) expression by increasing DNA methylation at its promoter, thereby dampening the migration and in vivo metastasis of gastric cancer cells. In addition, the levels of nuclear PCDH9 correlate with CDH2 expression, lymph node metastasis, and the prognosis of gastric cancer patients. Our finding demonstrates a unique mechanism of nuclear PCDH9-impaired gastric cancer metastasis by promoting DNA methylation of CDH2 promoter.

SELECTION OF CITATIONS
SEARCH DETAIL