Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
2.
Gastric Cancer ; 26(1): 69-81, 2023 01.
Article in English | MEDLINE | ID: mdl-36114400

ABSTRACT

BACKGROUND: Ubiquitous mitochondrial creatine kinase (uMtCK) transfers high-energy phosphates from mitochondrially generated ATP to creatine to generate phosphocreatine. uMtCK overexpression has been reported in several malignant tumors, however, the clinical significance and impact of uMtCK in gastric cancer (GC) has not been comprehensively studied. METHODS: We first examined uMtCK expression in GC by quantitative real-time PCR and western blot assays. Then the clinicopathological significance of aberrant uMtCK expression was determined by immunohistochemical staining in a GC tissue microarray. Kaplan-Meier analysis was used for survival analysis. The biological functions of uMtCK in GC cells were explored by wound-healing, transwell assays and glucose metabolism assays in vitro as well as a liver metastasis model by spleen injection in nude mice in vivo. RESULTS: We verified that the expression of uMtCK was substantially elevated in GC tissues, significantly associating with a poorer prognosis in GC patients, especially for those with advanced stage. In univariate and multivariate analyses, uMtCK expression emerged as an independent prognostic factor for both disease-free survival and overall survival. Functionally, we demonstrated that uMtCK promoted glycolysis in GC cells and facilitated their migration, invasion and liver metastasis in vitro and in vivo. Mechanistically, uMtCK enhanced GC progression in a HK2-dependent glycolysis via acting the JNK-MAPK/JUN signaling pathway. CONCLUSIONS: uMtCK could serve as a novel independent prognostic biomarker as well as potential therapeutic target for GC patients, particularly for GC patients with an advanced UICC stage and tumor recurrence.


Subject(s)
Liver Neoplasms , Stomach Neoplasms , Mice , Animals , Humans , Stomach Neoplasms/pathology , Creatine Kinase, Mitochondrial Form/metabolism , Mice, Nude , Glycolysis , Cell Proliferation , Prognosis , Gene Expression Regulation, Neoplastic , Cell Line, Tumor
3.
J Extracell Vesicles ; 11(1): e12186, 2022 01.
Article in English | MEDLINE | ID: mdl-35041299

ABSTRACT

Liver metastasis of colorectal cancer (CRLM) is the most common cause of CRC-related mortality, and is typically caused by interactions between CRC cells and the tumour microenvironment (TME) in the liver. However, the molecular mechanisms underlying the crosstalk between tumour-derived extracellular vesicle (EV) miRNAs and the TME in CRLM have yet to be fully elucidated. The present study demonstrated that highly metastatic CRC cells released more miR-181a-5p-rich EVs than cells which exhibit a low metastatic potential, in-turn promoting CRLM. Additionally, we verified that FUS mediated packaging of miR-181a-5p into CRC EVs, which in-turn persistently activated hepatic stellate cells (HSCs) by targeting SOCS3 and activating the IL6/STAT3 signalling pathway. Activated HSCs could secrete the chemokine CCL20 and further activate a CCL20/CCR6/ERK1/2/Elk-1/miR-181a-5p positive feedback loop, resulting in reprogramming of the TME and the formation of pre-metastatic niches in CRLM. Clinically, high levels of serum EV containing miR-181a-5p was positively correlated with liver metastasis in CRC patients. Taken together, highly metastatic CRC cells-derived EVs rich in miR-181a-5p could activate HSCs and remodel the TME, thereby facilitating liver metastasis in CRC patients. These results provide novel insight into the mechanism underlying liver metastasis in CRC.


Subject(s)
Colorectal Neoplasms/pathology , Extracellular Vesicles/metabolism , Hepatic Stellate Cells/metabolism , Liver Neoplasms/secondary , MicroRNAs/metabolism , Tumor Microenvironment , Biomarkers, Tumor/metabolism , Chemokine CCL20/metabolism , Colorectal Neoplasms/metabolism , Feedback, Physiological , Humans , Interleukin-6/metabolism , Liver/metabolism , Liver Neoplasms/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , RNA-Binding Protein FUS/metabolism , Receptors, CCR6/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/metabolism , ets-Domain Protein Elk-1/metabolism
4.
Oncogene ; 40(28): 4709-4724, 2021 07.
Article in English | MEDLINE | ID: mdl-34145399

ABSTRACT

Glycolysis plays a crucial role in reprogramming the metastatic tumor microenvironment. A series of lncRNAs have been identified to function as oncogenic molecules by regulating glycolysis. However, the roles of glycolysis-related lncRNAs in regulating colorectal cancer liver metastasis (CRLM) remain poorly understood. In the present study, the expression of the glycolysis-related lncRNA MIR17HG gradually increased from adjacent normal to CRC to the paired liver metastatic tissues, and high MIR17HG expression predicted poor survival, especially in patients with liver metastasis. Functionally, MIR17HG promoted glycolysis in CRC cells and enhanced their invasion and liver metastasis in vitro and in vivo. Mechanistically, MIR17HG functioned as a ceRNA to regulate HK1 expression by sponging miR-138-5p, resulting in glycolysis in CRC cells and leading to their invasion and liver metastasis. More interestingly, lactate accumulated via glycolysis activated the p38/Elk-1 signaling pathway to promote the transcriptional expression of MIR17HG in CRC cells, forming a positive feedback loop, which eventually resulted in persistent glycolysis and the invasion and liver metastasis of CRC cells. In conclusion, the present study indicates that the lactate-responsive lncRNA MIR17HG, acting as a ceRNA, promotes CRLM through a glycolysis-mediated positive feedback circuit and might be a novel biomarker and therapeutic target for CRLM.


Subject(s)
Liver Neoplasms , Glycolysis , RNA, Long Noncoding
6.
J Hematol Oncol ; 13(1): 156, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33213490

ABSTRACT

BACKGROUND: Mounting evidence has demonstrated the vital importance of tumor-associated macrophages (TAMs) and exosomes in the formation of the premetastatic niche. However, the molecular mechanisms by which tumor-derived exosomal miRNAs interact with TAMs underlying premetastatic niche formation and colorectal cancer liver metastasis (CRLM) remain largely unknown. METHODS: Transmission electron microscopy and differential ultracentrifugation were used to verify the existence of exosomes. In vivo and in vitro assays were used to identify roles of exosomal miR-934. RNA pull-down assay, dual-luciferase reporter assay, etc. were applied to clarify the mechanism of exosomal miR-934 regulated the crosstalk between CRC cells and M2 macrophages. RESULTS: In the present study, we first demonstrated the aberrant overexpression of miR-934 in colorectal cancer (CRC), especially in CRLM, and its correlation with the poor prognosis of CRC patients. Then, we verified that CRC cell-derived exosomal miR-934 induced M2 macrophage polarization by downregulating PTEN expression and activating the PI3K/AKT signaling pathway. Moreover, we revealed that hnRNPA2B1 mediated miR-934 packaging into exosomes of CRC cells and then transferred exosomal miR-934 into macrophages. Interestingly, polarized M2 macrophages could induce premetastatic niche formation and promote CRLM by secreting CXCL13, which activated a CXCL13/CXCR5/NFκB/p65/miR-934 positive feedback loop in CRC cells. CONCLUSIONS: These findings indicate that tumor-derived exosomal miR-934 can promote CRLM by regulating the crosstalk between CRC cells and TAMs. These findings reveal a tumor and TAM interaction in the metastatic microenvironment mediated by tumor-derived exosomes that affects CRLM. The present study also provides a theoretical basis for secondary liver cancer.


Subject(s)
Colorectal Neoplasms/pathology , Liver Neoplasms/secondary , Macrophage Activation , MicroRNAs/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Exosomes/genetics , Exosomes/immunology , Exosomes/pathology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Macrophages/immunology , Macrophages/pathology , MicroRNAs/genetics , Up-Regulation
7.
Cell Death Dis ; 11(3): 206, 2020 03 30.
Article in English | MEDLINE | ID: mdl-32231199

ABSTRACT

Gastric cancer (GC) has a high mortality rate, and metastasis is the main reason for treatment failure. It is important to study the mechanism of tumour invasion and metastasis based on the regulation of key genes. In a previous study comparing the expression differences between GES-1 and SGC-7901 cells, PCDHGA9 was selected for further research. In vitro and in vivo experiments showed that PCDHGA9 inhibited invasion and metastasis. A cluster analysis suggested that PCDHGA9 inhibited epithelial-mesenchymal transition (EMT) through the Wnt/ß-catenin and TGF-ß pathways. Laser confocal techniques and western blotting revealed that PCDHGA9 inhibited the nuclear translocation of ß-catenin, regulated T cell factor (TCF)/ /lymphoid enhancer factor (LEF) transcriptional activity, directly impacted the signal transmission of the TGF-ß/Smad2/3 pathway, strengthened the adhesion complex, weakened the effects of TGF-ß, and blocked the activation of the Wnt pathway. In addition, PCDHGA9 expression was regulated by methylation, which was closely related to poor clinical prognosis. The aim of this study was to elucidate the molecular mechanism by which PCDHGA9 inhibits EMT and metastasis in GC to provide a new theoretical basis for identifying GC metastasis and a new target for improving the outcome of metastatic GC.


Subject(s)
Cadherins/genetics , Stomach Neoplasms/genetics , beta Catenin/genetics , Aged , Animals , Cadherins/metabolism , Epithelial-Mesenchymal Transition , Female , Genes, Tumor Suppressor , Heterografts , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Metastasis , Protocadherins , Stomach Neoplasms/metabolism , Transcription, Genetic , beta Catenin/metabolism
8.
Biochem Biophys Res Commun ; 505(3): 816-822, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30297112

ABSTRACT

The lack of efficient tumor invasion and metastatic biomarkers led to high mortality rates in colon cancer patients. Aberrant expression of ubiquitin-specific protease 6 (USP6) was involved in several diseases including cancer, while its role in the progression of colon cancer was still unclear. In this study, USP6 was evaluated at both mRNA and protein levels by using RT-PCR, western blot and immunohistochemistry staining analyses. The results revealed that high USP6 expression predicted poor disease-specific survival and overall survival through Kaplan-Meier analyses with log-rank tests, univariate and multivariate Cox analyses. Furthermore, cell function assay demonstrated that USP6 could promote colon cancer cells' invasion in vitro and liver metastasis in vivo. These findings indicated that high USP6 expression contributed to the progression of colon cancer and USP6 may be a valuable prognostic factor in patients with colon cancer.


Subject(s)
Colonic Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Proto-Oncogene Proteins/physiology , Ubiquitin Thiolesterase/physiology , Aged , Cell Line, Tumor , Colonic Neoplasms/diagnosis , Colonic Neoplasms/mortality , Disease Progression , Female , Humans , Liver Neoplasms/pathology , Male , Middle Aged , Prognosis , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics , RNA, Messenger/analysis , Survival Analysis , Ubiquitin Thiolesterase/analysis , Ubiquitin Thiolesterase/genetics
9.
Cell Death Dis ; 9(2): 27, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348665

ABSTRACT

The results of a cDNA  array revealed that protocadherin gamma subfamily A, 9 (PCDHGA9) was significantly decreased in SGC-7901 gastric cancer (GC) cells compared with GES-1 normal gastric cells and was strongly associated with the Wnt/ß-catenin and transforming growth factor-ß (TGF-ß)/Smad2/3 signaling pathway. As a member of the cadherin family, PCDHGA9 functions in both cell-cell adhesion and nuclear signaling. However, its role in tumorigenicity or metastasis has not been reported. In the present study, we found that PCDHGA9 was decreased in GC tissues compared with corresponding normal mucosae and its expression was correlated with the GC TNM stage, the UICC stage, differentiation, relapse, and metastasis (p < 0.01). Multivariate Cox analysis revealed that PCDHGA9 was an independent prognostic indicator for overall survival (OS) and disease-free survival (DFS) (p < 0.01). The effects of PCDHGA9 on GC tumor growth and metastasis were examined both in vivo and in vitro. PCDHGA9 knockdown promoted GC cell proliferation, migration, and invasion, whereas PCDHGA9 overexpression inhibited GC tumor growth and metastasis but induced apoptosis, autophagy, and G1 cell cycle arrest. Furthermore, PCDHGA9 suppressed epithelial-mesenchymal transition (EMT) induced by TGF-ß, decreased the phosphorylation of Smad2/3, and inhibited the nuclear translocation of pSmad2/3. Our results suggest that PCDHGA9 might interact with ß-catenin to prevent ß-catenin from dissociating in the cytoplasm and translocating to the nucleus. Moreover, PCDHGA9 overexpression restrained cell proliferation and reduced the nuclear ß-catenin, an indicator of Wnt/ß-catenin pathway activation, suggesting that PCDHGA9 negatively regulates Wnt signaling. Together, these data indicate that PCDHGA9 acts as a tumor suppressor with anti-proliferative activity and anti-invasive ability, and the reduction of PCDHGA9 could serve as an independent prognostic biomarker in GC.


Subject(s)
Cadherins/genetics , Stomach Neoplasms/genetics , Aged , Apoptosis , Autophagy , Cadherin Related Proteins , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Genes, Tumor Suppressor , Humans , Male , Stomach Neoplasms/pathology
10.
Drug Deliv ; 24(1): 443-451, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28165858

ABSTRACT

This article reports a promising approach to enhance the oral delivery of nuciferine (NUC), improve its aqueous solubility and bioavailability, and allow its controlled release as well as inhibiting lipid accumulation. NUC-loaded poly lactic-co-glycolic acid nanoparticles (NUC-PLGA-NPs) were prepared according to a solid/oil/water (s/o/w) emulsion technique due to the water-insolubility of NUC. PLGA exhibited excellent loading capacity for NUC with adjustable dosing ratios. The drug loading and encapsulation efficiency of optimized formulation were 8.89 ± 0.71 and 88.54 ± 7.08%, respectively. NUC-PLGA-NPs exhibited a spherical morphology with average size of 150.83 ± 5.72 nm and negative charge of -22.73 ± 1.63 mV, which are suitable for oral administration. A sustained NUC released from NUC-PLGA-NPs with an initial exponential release owing to the surface associated drug followed by a slower release of NUC, which was entrapped in the core. In addition, ∼77 ± 6.67% was released in simulating intestinal juice, while only about 45.95 ± 5.2% in simulating gastric juice. NUC-PLGA-NPs are more efficient against oleic acid (OA)-induced hepatic steatosis in HepG2 cells when compared to naked NUC (n-NUC, *p < 0.05). The oral bioavailability of NUC-PLGA-NPs group was significantly higher (**p < 0.01) and a significantly decreased serum levels of total cholesterol (TC), triglycerides (TG) and low-density lipoprotein cholesterol (LDL-C), as well as a higher concentration of high-density lipoprotein cholesterol (HDL-C) was observed, compared with that of n-NUC treated group. These findings suggest that NUC-PLGA-NPs hold great promise for sustained and controlled drug delivery with improved bioavailability to alleviating lipogenesis.


Subject(s)
Aporphines/chemical synthesis , Drug Delivery Systems/methods , Fatty Liver/drug therapy , Lactic Acid/chemical synthesis , Nanoparticles/chemistry , Polyglycolic Acid/chemical synthesis , Administration, Oral , Animals , Aporphines/administration & dosage , Aporphines/metabolism , Chemical Phenomena , Fatty Liver/metabolism , Hep G2 Cells , Humans , Lactic Acid/administration & dosage , Lactic Acid/metabolism , Male , Nanoparticles/administration & dosage , Nanoparticles/metabolism , Polyglycolic Acid/administration & dosage , Polyglycolic Acid/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer , Random Allocation , Rats , Rats, Sprague-Dawley
11.
Mol Cancer ; 16(1): 12, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28095858

ABSTRACT

BACKGROUND: Despite advancements in the diagnosis and treatment of colorectal cancer (CRC), many patients die because of tumor metastasis or recurrence. Therefore, identifying new prognostic markers and elucidating the mechanisms of CRC metastasis and recurrence will help to improve the prognosis of the disease. As dysregulation of microRNAs is strongly related to cancer progression, the aim of this study was to identify the role of miR-4775 in the prognosis of CRC patients and the underling mechanisms involved in CRC progression. METHODS: qPCR and in situ hybridization were used to evaluate the expression of miR-4775 in 544 pairs of paraffin-embedded normal and CRC tissues. Kaplan-Meier analysis with the log-rank test was used for survival analyses. Immunohistochemical staining was applied to investigate the expression of miR-4775-regulated Smad7/TGFß pathway-associated markers. In vitro and in vivo invasion and metastasis assays were used to explore the function of miR-4775 in the progression of CRC. RESULTS: miR-4775 was identified as a high-risk factor for CRC metastasis and recurrence, with high levels predicting poor survival among the 544 studied CRC patients. Furthermore, high miR-4775 expression promoted the invasion of CRC cells as well as metastasis and the epithelial to mesenchymal transition (EMT) via Smad7-mediated activation of TGFß signaling both in vitro and in vivo. Downregulating miR-4775 or overexpressing Smad7 reversed the tumor-promoting roles of miR-4775/Smad7/TGFß in vitro and in vivo. CONCLUSION: miR-4775 promotes CRC metastasis and recurrence in a Smad7/TGFß signaling-dependent manner, providing a new therapeutic target for inhibiting the metastasis or recurrence of the disease.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Smad7 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease Models, Animal , Heterografts , Humans , Kaplan-Meier Estimate , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Prognosis , Signal Transduction
12.
Cancer Lett ; 389: 11-22, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28043911

ABSTRACT

We previously discovered that Ras association domain family member 6 (RASSF6) was downregulated and predicted poor prognosis in GC patients. However, the mechanisms of the down regulation of RASSF6 in GC remained unclear. Increasing evidence indicates that dysregulation of microRNAs promotes the progression of cancer through the repression of tumour suppressors. Here, we identified miR-181a-5p as a novel regulator of RASSF6 in GC. Functionally, ectopic expression or silencing of miR-181a-5p, respectively, promoted or inhibited GC cell proliferation, colony formation and cell cycle transition, as well as enhanced or prevented the invasion, metastasis of GC cells and epithelial to mesenchymal transition of GC cells in vitro and in vivo. Molecularly, miR-181a-5p functioned as an onco-miRNA by activating the RASSF6-regulated MAKP pathway. Overexpression or silencing of RASSF6 could partially reverse the effects of the overexpression or repression of miR-181a-5p on GC progress caused by activation of the MAKP pathway in vitro and in vivo. Clinically, high miR-181a-5p expression predicted poor survival in GC patients, especially combined with low RASSF6 expression. Collectively, we identified miR-181a-5p as an onco-miRNA, which acts by directly repressing RASSF6 in GC.


Subject(s)
MAP Kinase Signaling System/physiology , MicroRNAs/physiology , Monomeric GTP-Binding Proteins/physiology , Stomach Neoplasms/etiology , Apoptosis Regulatory Proteins , Cell Proliferation , Disease Progression , Epithelial-Mesenchymal Transition , Humans , Neoplasm Invasiveness , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology
13.
Am J Cancer Res ; 6(8): 1636-49, 2016.
Article in English | MEDLINE | ID: mdl-27648355

ABSTRACT

Serpina family A member 4 (SERPINA4), also known as kallistatin, exerts important effects in inhibiting tumor growth and angiogenesis in many malignancies. However, the precise role of SERPINA4 in CRC has not been fully elucidated. The present study aimed to investigate the expression of SERPINA4 and its clinical significance in CRC. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot analyses showed that the mRNA and protein expression of SERPINA4 in colorectal cancer (CRC) specimens was significantly decreased than that in adjacent normal mucosa. Immunohistochemistry (IHC) was conducted to characterize the expression pattern of SERPINA4 by using a tissue microarray (TMA) containing 327 archived paraffin-embedded CRC specimens. Statistical analyses revealed that decreased SERPINA4 expression was significantly associated with invasion depth, nodal involvement, distant metastasis, American Joint Committee on Cancer (AJCC) stage, and tumor differentiation. SERPINA4 was also an independent prognostic indicator of disease-free survival and overall survival in patients with CRC. Furthermore, the impact of altered SERPINA4 expression on CRC cells was analyzed with a series of in vitro and in vivo assays. The results demonstrated that SERPINA4 significantly inhibits malignant tumor progression and serves as a novel prognostic indicator and a potential therapeutic target for CRC.

14.
Tumour Biol ; 37(11): 14585-14594, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27614686

ABSTRACT

This study investigated the significance of La-related protein 1 (LARP1) in the development and progression of colorectal cancer (CRC). Quantitative real-time polymerase chain reaction and Western blot analyses were carried out to determine the mRNA and protein expression of LARP1 in CRC tumor tissues and paired adjacent normal mucosa. The expression of LARP1 was upregulated in CRC. Immunohistochemical analysis using tissue microarray was performed. A positive correlation between LARP1 and proliferating cell nuclear antigen (PCNA) in the area of proliferation was observed using the Spearman's correlation coefficient test (r = 0.332, P < 0.01). The elevated expression of LARP1 significantly correlated with T stage (P = 0.02), N stage (P = 0.006), M stage (P < 0.001), American Joint Committee on Cancer (AJCC) stage (P = 0.04), differentiation rank (P < 0.001), and PCNA level (P < 0.001). In addition, the inhibitory effect of LARP1 knockdown on CRC cell proliferation was demonstrated using Cell Counting Kit-8 (CCK8) and colony-forming cell (CFC) assays. Multivariate analysis showed that LARP1 was an independent prognostic factor for overall survival (OS; hazard rate (HR) = 0.244; 95 % confidence interval (CI), 0.078-0.769; P = 0.016) and disease-free survival (DFS; HR = 0.281; 95 % CI, 0.086-0.917; P = 0.035) in CRC patients. LARP1 plays an important role in the proliferation of colorectal cancer and represents a new prognostic indicator.


Subject(s)
Autoantigens/biosynthesis , Autoantigens/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Proliferating Cell Nuclear Antigen/biosynthesis , RNA, Messenger/biosynthesis , Ribonucleoproteins/biosynthesis , Ribonucleoproteins/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/biosynthesis , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/mortality , Disease-Free Survival , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Staging , Prognosis , RNA Interference , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Tissue Array Analysis , SS-B Antigen
15.
Oncotarget ; 7(37): 60593-60608, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27533254

ABSTRACT

Barx2 is a Bar family homeodomain transcription factor shown to play a critical role in cell adhesion and cytoskeleton remodeling, key processes in carcinogenesis and metastasis. Using quantitative real-time PCR, Western blotting, and immunohistochemistry, we found that Barx2 is expressed at lower levels in human gastric cancer (GC) tissues than in adjacent normal mucosa. In a multivariate analysis, Barx2 expression emerged as an independent prognostic factor for disease-free and overall survival. Kaplan-Meier survival analysis showed a trend toward even shorter overall survival in the patient group with Barx2-negative tumors, independent of advanced UICC stage and tumor relapse. Using in vitro and in vivo assays, we demonstrated that under normal conditions Barx2 inhibited GC cell proliferation and invasiveness through inhibition of the Wnt/ß-catenin signaling pathway. These findings indicate that reduction or loss of Barx2 dis-inhibits GC cell proliferation and invasion, and that reduction in Barx2 could serve as an independent prognostic biomarker for poor outcome in GC patients.


Subject(s)
Biomarkers, Tumor/metabolism , Homeodomain Proteins/metabolism , Stomach Neoplasms/genetics , Aged , Animals , Biomarkers, Tumor/genetics , Carcinogenesis , Cell Adhesion , Cell Proliferation , Cytoskeleton/metabolism , Female , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , Male , Mice , Mice, Nude , Neoplasm Metastasis , Patient Outcome Assessment , Prognosis , Stomach Neoplasms/diagnosis , Stomach Neoplasms/mortality , Survival Analysis , Tissue Array Analysis , Xenograft Model Antitumor Assays
16.
Biochem Biophys Res Commun ; 478(1): 67-73, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27453340

ABSTRACT

Human BarH-like homeobox 2 (Barx2), a homeodomain factor of the Bar family, has an important role in controlling the expression of cell adhesion molecules and has been reported in an increasing array of tumor types except colorectal cancer (CRC). The purpose of the current study was to characterize the expression of Barx2 and assess the clinical significance of Barx2 in CRC. First, we analyzed the expression of Barx2 in two independent public datasets from Oncomine. Subsequently, we evaluated Barx2 mRNA and protein expression by quantitative real-time PCR and western blotting, respectively. It was determined that Barx2 expression was lower in tumor tissues than in adjacent non-tumorous colorectal tissues of CRC patients, consistent with results from the public datasets. Subsequently, a tissue microarray containing 196 CRC specimens was evaluated for Barx2 expression by immunohistochemical staining. It was found that low expression of Barx2 significantly correlated with TNM stage, AJCC stage, differentiation, and relapse in patients with CRC. Patients with lower levels of Barx2 expression showed reduced disease-free survival and overall survival. Furthermore, a trend toward shorter overall survival in the patient group with Barx2-negative tumors independent of advanced AJCC stage and poor differentiation was determined by Kaplan-Meier survival analysis. Based on univariate and multivariate analyses, Barx2 expression was an independent prognostic factor for determining CRC prognosis. Taken together, low Barx2 expression was associated with the progression of CRC and could serve as a potential independent prognostic biomarker for patients with CRC.


Subject(s)
Colon/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Down-Regulation , Homeodomain Proteins/genetics , Rectum/pathology , Aged , Colon/metabolism , Colorectal Neoplasms/diagnosis , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/analysis , Humans , Male , Middle Aged , Prognosis , Rectum/metabolism , Survival Analysis
17.
Oncotarget ; 7(15): 19794-812, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26942699

ABSTRACT

Conventional high-recurrence risk factors are not sufficient to predict post-operative risk of tumor recurrence or sensitivity to 5-fluorouracil (5-FU)-based chemotherapy for stage II colon cancer. DDA1, an evolutionarily conserved gene located at 19p13.11, may be involved in the activation of nuclear factor kappaB (NFκB). This study aimed to investigate whether DDA1 contributes to tumorigenesis and progression of stage II colon cancer via activation of the NFκB pathway. We found that positive expression of DDA1 alone or in combination with p65 nuclear translocation correlated with increased risk of tumor recurrence in patients with stage IIB-IIC colon cancer. DDA1 overexpression in colon cancer lines promoted cell proliferation, facilitated cell cycle progression, inhibited 5-FU-induced apoptosis, enhanced invasion, and induced the epithelial-mesenchymal transition. Suppression of DDA1 inhibited tumor progression, and reduced tumor growth in vivo. We also demonstrated that DDA1-mediated tumor progression is associated with the activation of the NFκB/COP9 signalosome 2(CSN2)/glycogen synthase kinase3ß (GSK3ß) pathway. These results indicate that DDA1 promotes colon cancer progression through activation of NFκB/CSN2/GSK3ß signaling. DDA1, together with NFκB activation status, may serve as a sensitive biomarker for tumor recurrence risk and prognosis in patients with stage IIB-IIC colon cancers.


Subject(s)
Colonic Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blotting, Western , COP9 Signalosome Complex/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , DNA-Binding Proteins/genetics , Disease Progression , Female , Fluorouracil/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , HCT116 Cells , HT29 Cells , Humans , Male , Mice, Nude , Middle Aged , NF-kappa B/metabolism , Neoplasm Staging , Postoperative Period , Signal Transduction/drug effects , Signal Transduction/genetics , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...