Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Mater Today Bio ; 26: 101096, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38831909

ABSTRACT

Conventional implantable electronics based on von Neumann architectures encounter significant limitations in computing and processing vast biological information due to computational bottlenecks. The memristor with integrated memory-computing and low power consumption offer a promising solution to overcome the computational bottleneck and Moore's law limitations of traditional silicon-based implantable devices, making them the most promising candidates for next-generation implantable devices. In this work, a highly stable memristor with an Ag/BaTiO3/MnO2/FTO structure was fabricated, demonstrating retention characteristics exceeding 1200 cycles and endurance above 1000 s. The device successfully exhibited three-stage responses to biological signals after implantation in SD (Sprague-Dawley) rats. Importantly, the memristor perform remarkable reversibility, maintaining over 100 cycles of stable repetition even after extraction from the rat. This study provides a new perspective on the biomedical application of memristors, expanding the potential of implantable memristive devices in intelligent medical fields such as health monitoring and auxiliary diagnostics.

3.
Pathol Res Pract ; 245: 154422, 2023 May.
Article in English | MEDLINE | ID: mdl-37003132

ABSTRACT

BACKGROUND: Hypoxia critically drives malignant tumor development and is characteristic of hepatocellular carcinoma (HCC), where HIF-1α plays a crucial role. The ubiquitin-conjugating enzyme E2K (UBE2K) is known to participate in the advancement of several human cancers. However, the role of UBE2K in HCC or whether it is a hypoxia-responsive gene remains to be further identified. METHOD: We performed a microarray to measure the gene expression differences between normoxia and hypoxia. CoCl2 mimicked the hypoxic condition. The protein and RNA expression of HIF-1α, UBE2K, and Actin in HCC cells were measured by western blotting(WB) and RT-qPCR, respectively. Immunohistochemical (IHC) staining analyzed the expression of UBE2K and HIF-1α in HCC tissues. CCK-8 and colony formation assay evaluated the HCC cell growth. Scratch healing and transwell assays were used to detect the migration capability of the cells. Lipofectamine 3000 was used to transfect the plasmids or siRNAs to HCC cells. RESULTS: We identified UBE2K as a potential hypoxia-responsive gene. Our study showed that hypoxia induced HIF-1α-mediated increase of UBE2K levels in HCC cells, which decreased under HIF-1α deficiency under hypoxia. Further bioinformatics analysis based on UALCAN and GEPIA databases confirmed that UBE2K was highly expressed in HCC tissues and positively associated with HIF-1α expression. Functionally, Hep3B and Huh7 cell proliferation and migration were stimulated upon UBE2K overexpression, while the UBE2K knockdown suppressed such effect. Furthermore, functional rescue experiment proved that depletion of UBE2K inhibited hypoxia-induced cell proliferation and migration in HCC cells. In contrast, enhancing UBE2K levels rescued cell proliferation and migration repression caused by HIF-1α deficiency in hypoxia. CONCLUSION: Our results established UBE2K as a potential hypoxia-inducible gene in HCC cells, positively regulated by HIF-1α in hypoxia. Moreover, UBE2K served as an oncogene and cooperated with HIF-1α to form a functional HIF-1α/UBE2K axis to trigger HCC progression, highlighting a potential application of UBE2K as a therapeutic target for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Cell Line, Tumor , Hypoxia , Cell Hypoxia , Cell Proliferation/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Gene Expression Regulation, Neoplastic/genetics , Cell Movement/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
4.
Biochem Biophys Res Commun ; 638: 210-218, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36481361

ABSTRACT

Hepatocellular carcinoma (HCC) is a serious threat to human health and life due to its high morbidity and mortality. Ubiquitin-conjugating enzymes are players in the ubiquitin proteasome system and are responsible for a great number of physiological activities in cells. The action of ubiquitin-conjugating enzyme UBE2K in HCC has not been reported. Therefore, we studied the function and role of UBE2K in the malignant progression of HCC. An analysis of UBE2K expression in HCC cells was performed using RT-qPCR and protein immunoblotting. CCK-8, Transwell and sphere formation assays were used to identify the potential effects of UBE2K in HCC cell proliferation, migration and stemness property. RT-qPCR, and protein immunoblotting experiments was taken to explore the regulation between UBE2K and c-Myc. Here, we discovered that UBE2K expression was elevated in HCC cells, and elevated UBE2K predicts worse prognosis for HCC patients. Functionally, UBE2K promote, while UBE2K knockdown suppressed cell proliferation, migration and stemness property of HCC cells. Furthermore, c-Myc was identified as a downstream target of UBE2K. Moreover, functional rescue experiments finally proved that UBE2K facilitates the malignant progression of HCC cells by upregulating c-Myc. We clarified through in vivo experiments that UBE2K expression promotes tumor growth in HCC. Taken together, our study results proved the molecular regulation of UBE2K and c-Myc in HCC and the oncogenic role of UBE2K/c-Myc axis in HCC progression, thus it provides a promising molecular target for the diagnosis and treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Cell Line , Cell Proliferation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
6.
J Cancer ; 13(11): 3177-3188, 2022.
Article in English | MEDLINE | ID: mdl-36118523

ABSTRACT

The hypoxic tumor microenvironment, a fundamental feature of solid tumors, drives hepatocellular carcinoma (HCC) progression through regulating the transcriptional activities of protein-coding and noncoding genes. However, long noncoding RNA (lncRNA)-mediated HCC progression in hypoxic microenvironment remains largely unknown yet. In this study, we found that LINC00674 was upregulated under hypoxic conditions in a HIF-1-dependent manner, and the occupancy of HIF-1 to HRE of LINC00674 gene promoter was essential for its transcription. In addition, LINC00674 level was increased in HCC cell lines and tissues. Clinically, statistical analysis showed that LINC00674 expression was significantly associated with tumor size, venous infiltration, tumor stage and poor prognosis of HCC. Functionally, loss-of-function assays revealed that LINC00674 knockdown inhibited the migration, proliferation and invasion of HCC cells. Furthermore, LINC00674 silencing prominently repressed the mTOR signaling pathway. LINC00674 overexpression-enhanced HCC cell proliferation, migration and invasion were markedly abolished by an mTOR inhibitor rapamycin. NADPH oxidase 1 (NOX1) was positively regulated by LINC00674 in HCC cells. NOX1 knockdown markedly reversed LINC00674-upregulated the p-mTOR level and HCC cells' malignant behaviors. Finally, we found that LINC00674 knockdown attenuated the growth of HCC cells in vivo. Our finding demonstrated that LINC00674 was a new HIF-1 target gene, and hypoxia-induced LINC00674 exerted a pro-proliferative and pro-metastatic role in HCC, possibly by activating the NOX1/mTOR signaling pathway. This study suggested LINC00674 as a promising therapeutic target for HCC.

8.
Int J Med Sci ; 18(16): 3749-3758, 2021.
Article in English | MEDLINE | ID: mdl-34790050

ABSTRACT

The ubiquitin-conjugating enzyme (E2) is a critical component of the ubiquitin-proteasome system and regulates hepatocarcinogenesis by controlling protein degradation. Ubiquitin-conjugating enzyme E2 O (UBE2O), a member of the E2 family, functions as an oncogene in human cancers. Nevertheless, the role of UBE2O in hepatocellular carcinoma (HCC) remains unknown yet. Here, we demonstrated that the UBE2O level was markedly upregulated in HCC compared with adjacent noncancerous tissues. UBE2O overexpression was also confirmed in HCC cell lines. UBE2O overexpression was prominently associated with advanced tumor stage, high tumor grade, venous infiltration, and reduced HCC patients' survivals. UBE2O knockdown inhibited the migration, invasion, and proliferation of HCCLM3 cells. UBE2O overexpression enhanced the proliferation and mobility of Huh7 cells. Mechanistically, UBE2O mediated the ubiquitination and degradation of AMP-activated protein kinase α2 (AMPKα2) in HCC cells. UBE2O silencing prominently increased AMPKα2 level and reduced phosphorylated mechanistic target of rapamycin kinase (p-mTOR), MYC, Cyclin D1, HIF1α, and SREBP1 levels in HCCLM3 cells. UBE2O depletion markedly activated the AMPKα2/mTOR pathway in Huh7 cells. Moreover, AMPKα2 silencing reversed UBE2O downregulation-induced mTOR pathway inactivation. Rapamycin, an inhibitor of mTOR, remarkably abolished UBE2O-induced mTOR phosphorylation and HCC cell proliferation and mobility. To conclude, UBE2O was highly expressed in HCC and its overexpression conferred to the poor clinical outcomes of patients. UBE2O contributed to the malignant behaviors of HCC cells, including cell proliferation, migration, and invasion, by reducing AMPKα2 stability and activating the mTOR pathway.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Ubiquitin-Conjugating Enzymes/physiology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adult , Aged , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Male , Middle Aged , Neoplasm Invasiveness , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
9.
J Cancer ; 12(22): 6805-6813, 2021.
Article in English | MEDLINE | ID: mdl-34659569

ABSTRACT

Long non-coding RNAs (lncRNAs) are critical drivers and suppressors of human hepatocellular carcinoma (HCC). The downregulation of transmembrane protein 220 antisense RNA 1 (TMEM220-AS1) is correlated with poor prognosis in HCC. Nevertheless, the role of TMEM220-AS1 in HCC and the underlying mechanism remains unclear. In this study, TMEM220-AS1 levels were markedly reduced in HCC tissues compared with noncancerous tissues. TMEM220-AS1 downregulation was confirmed in HCC cell lines. TMEM220-AS1 expression was associated with tumor stage, venous infiltration, tumor size, and survival of HCC patients. TMEM220-AS1 overexpression suppressed the migration, invasion, and proliferation of HCC cells. Interestingly, ectopic expression of TMEM220-AS1 increased TMEM220 levels in HCC cells. Decreased TMEM220 levels were observed in HCC tissues and cell lines. TMEM220 expression was positively correlated with TMEM220-AS1 levels in HCC tissue samples and TMEM220 downregulation was significantly correlated with reduced patient survival. TMEM220 overexpression suppressed HCC cell proliferation and mobility. TMEM220 knockdown eliminated the suppressive effect of TMEM220-AS1 in HCCLM3 cells. Mechanistically, TMEM220 overexpression reduced the nuclear accumulation of ß-catenin and decreased MYC, Cyclin D1, and Snail1 mRNA levels in HCCLM3 cells. BIO, a GSK3ß inhibitor, eliminated TMEM220-induced Wnt/ß-catenin pathway inactivation and inhibited HCC cell proliferation and mobility. In conclusion, TMEM220-AS1 and TMEM220 were expressed at low levels in HCC patients. TMEM220-AS1 inhibited the malignant behavior of HCC cells by enhancing TMEM220 expression and subsequently inactivating the Wnt/ß-catenin pathway.

10.
EXCLI J ; 20: 537-549, 2021.
Article in English | MEDLINE | ID: mdl-33883982

ABSTRACT

This study aimed to evaluate the safety and efficacy of rivaroxaban in preventing portal vein system thrombosis (PVST) in patients with liver cirrhosis after splenectomy and pericardial devascularization. 70 cirrhotic patients undergoing splenectomy and pericardial devascularization were randomly assigned to rivaroxaban treatment (n=35) or low-molecular weight heparin (LMWH) plus warfarin treatment (n=35) for 30 days in this randomized controlled trial. The primary endpoint is the PVST formation. Ultrasound doctors and radiologists were blinded to the randomization results. Both groups received routine outpatient inspection every month and were followed for one year. 17 patients (48.6 %) in rivaroxaban group developed PVST, compared with 27 patients (77.1 %) in LMWH plus warfarin group (P=0.025). The incidence of PVST during the first year postoperation was significantly lower in rivaroxaban group than in LMWH plus warfarin group (F=7.901, P=0.006). The intra-group comparisons versus baseline showed the liver function improved from POD 21 to POM 1, and coagulation function improved from POM 2, in rivaroxaban group. In contrast, the liver function improved from POM 1 to POM 2, and coagulation function improved from POM 4, in LMWH plus warfarin group. The prophylactic use of rivaroxaban significantly decreases the incidence of PVST after splenectomy and pericardial devascularization in cirrhotic patients compared to LMWH plus warfarin treatment. Besides, rivaroxaban treatment was safe and effective and associated with better liver and coagulation functions improvement than LMWH plus warfarin treatment.

11.
BMC Bioinformatics ; 20(Suppl 22): 720, 2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31888439

ABSTRACT

BACKGROUND: Portal vein system thrombosis (PVST) is potentially fatal for patients if the diagnosis is not timely or the treatment is not proper. There hasn't been any available technique to detect clinic risk factors to predict PVST after splenectomy in cirrhotic patients. The aim of this study is to detect the clinic risk factors of PVST for splenectomy and cardia devascularization patients for liver cirrhosis and portal hypertension, and build an efficient predictive model to PVST via the detected risk factors, by introducing the machine learning method. We collected 92 clinic indexes of splenectomy plus cardia devascularization patients for cirrhosis and portal hypertension, and proposed a novel algorithm named as RFA-PVST (Risk Factor Analysis for PVST) to detect clinic risk indexes of PVST, then built a SVM (support vector machine) predictive model via the detected risk factors. The accuracy, sensitivity, specificity, precision, F-measure, FPR (false positive rate), FNR (false negative rate), FDR (false discovery rate), AUC (area under ROC curve) and MCC (Matthews correlation coefficient) were adopted to value the predictive power of the detected risk factors. The proposed RFA-PVST algorithm was compared to mRMR, SVM-RFE, Relief, S-weight and LLEScore. The statistic test was done to verify the significance of our RFA-PVST. RESULTS: Anticoagulant therapy and antiplatelet aggregation therapy are the top-2 risk clinic factors to PVST, followed by D-D (D dimer), CHOL (Cholesterol) and Ca (calcium). The SVM (support vector machine) model built on the clinic indexes including anticoagulant therapy, antiplatelet aggregation therapy, RBC (Red blood cell), D-D, CHOL, Ca, TT (thrombin time) and Weight factors has got pretty good predictive capability to PVST. It has got the highest PVST predictive accuracy of 0.89, and the best sensitivity, specificity, precision, F-measure, FNR, FPR, FDR and MCC of 1, 0.75, 0.85, 0.92, 0, 0.25, 0.15 and 0.8 respectively, and the comparable good AUC value of 0.84. The statistic test results demonstrate that there is a strong significant difference between our RFA-PVST and the compared algorithms, including mRMR, SVM-RFE, Relief, S-weight and LLEScore, that is to say, the risk indicators detected by our RFA-PVST are statistically significant. CONCLUSIONS: The proposed novel RFA-PVST algorithm can detect the clinic risk factors of PVST effectively and easily. Its most contribution is that it can display all the clinic factors in a 2-dimensional space with independence and discernibility as y-axis and x-axis, respectively. Those clinic indexes in top-right corner of the 2-dimensional space are detected automatically as risk indicators. The predictive SVM model is powerful with the detected clinic risk factors of PVST. Our study can help medical doctors to make proper treatments or early diagnoses to PVST patients. This study brings the new idea to the study of clinic treatment for other diseases as well.


Subject(s)
Cardia/pathology , Hypertension, Portal/complications , Liver Cirrhosis/complications , Portal Vein/pathology , Splenectomy/adverse effects , Venous Thrombosis/diagnosis , Venous Thrombosis/etiology , Algorithms , Area Under Curve , Humans , Liver Cirrhosis/pathology , Postoperative Complications/diagnosis , Postoperative Complications/etiology , Reproducibility of Results , Risk Factors
12.
J Exp Clin Cancer Res ; 37(1): 231, 2018 Sep 19.
Article in English | MEDLINE | ID: mdl-30231922

ABSTRACT

BACKGROUND: Cartilage oligomeric matrix protein (COMP) is known to promote fibrosis in skin, lung and liver. Emerging evidence shows that COMP plays critical roles in tumor development, including breast cancer, colon cancer and hepatocellular carcinoma (HCC). Nevertheless, the role of COMP in HCC proliferation and metastasis and its underlying mechanisms remain fully unclear. METHODS: Serum COMP was determined by ELISA. Cell Counting Kit-8 and plate colony formation were performed to evaluate cell proliferation. Wound healing and transwell assays were used to determine migration and invasion of HCC cells. Western blotting and immunofluorescence were carried out for detection of epithelial-to-mesenchymal transition (EMT) markers and MMPs in HCC cells. The in vivo role of COMP was evaluated using mouse models. We also measured effects of hepatic stellate cells (HSCs)-conditioned medium (CM) on HCC progression using transwell coculture system. RESULTS: Here, we found that serum COMP levels in HCC patients were significantly higher than those in healthy controls. Accordingly, high serum COMP levels in HCC patients significantly correlated with malignant clinical characteristics and poor clinical outcomes. Next, we investigated that recombinant human COMP protein (rCOMP) treatment resulted in increased abilities of proliferation, invasion and migration of HCC cells. Furthermore, rCOMP treatment enhanced proliferative and metastatic colonization of HCC cells in vivo. Mechanistically, CD36 receptor played an essential role in COMP-mediated HCC cell proliferation and metastasis. Functionally, COMP/CD36 signaling caused phosphorylation of ERK and AKT, resulting in the upregulation of tumor-progressive genes such as EMT markers, MMP-2/9, Slug and Twist in HCC cells. Interestingly, we revealed that COMP was secreted by HSCs. CM of LX2 cells with COMP knockdown showed weaker effects on the activation of MEK/ERK and PI3K/AKT signaling pathways in HCC cells compared to control CM. CONCLUSIONS: Our findings indicated that HSCs-derived COMP collaborated with CD36 and subsequently played an essential role in MEK/ERK and PI3K/AKT-mediated HCC progression. COMP might act as a promising target for the diagnosis and treatment of aggressive HCC.


Subject(s)
CD36 Antigens/genetics , Carcinoma, Hepatocellular/genetics , Cartilage Oligomeric Matrix Protein/genetics , Liver Neoplasms/genetics , Liver/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Cell Proliferation/genetics , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Hep G2 Cells , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver/pathology , Liver Neoplasms/pathology , MAP Kinase Signaling System/genetics , Mice , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics , Xenograft Model Antitumor Assays
13.
Oncotarget ; 8(50): 87737-87749, 2017 Oct 20.
Article in English | MEDLINE | ID: mdl-29152116

ABSTRACT

Recent studies have reported that microRNA-526b (miR-526b) is implicated in the growth and metastasis of cancer cells. However, the clinical significance of miR-526b and its role as well as underlying mechanisms are largely unknown in hepatocellular carcinoma (HCC). Here, we detected miR-526b expression difference between HCC and matched nontumor tissues with qRT-PCR. We found that miR-526b displayed lower expression in HCC patient tissues and cells. Clinical analysis revealed that low miR-526b expression correlated with large tumor size, venous infiltration, advanced tumor-node-metastasis (TNM) stage. Furthermore, miR-526b underexpression independently predicted poor prognosis of HCC patients. Functionally, we demonstrated that miR-526b inhibited proliferation, migration and invasion of HCC cells in vitro. Moreover, miR-526b overexpression restrained the tumor growth and pulmonary metastasis in vivo. Mechanistically, we proved that miR-526b could directly bind to 3'UTR of sirtuin 7 (SIRT7) mRNA and repressed its expression. miR-526b and SIRT7 showed a negative correlation in HCC tissues. More importantly, up-regulating SIRT7 expression antagonized miR-526b-inhibited proliferation, migration and invasion in SMMC-7721 cells. Furthermore, miR-526b suppressed epithelial-to-mesenchymal transition (EMT) of HCC cells. Immunoblotting analysis indicated that miR-526b reduced the levels of phosphorylated ERK (p-ERK), c-Myc, Cyclin D1, c-Jun, SNAIL and SLUG in HCC cells. SIRT7 restoration promoted phosphorylation of ERK and EMT in miR-526b overexpressing SMMC-7721 cells. Taken together, this is the first time we demonstrated that miR-526b might function as a prognostic biomarker and suppressed SIRT7 expression, and subsequently led to the growth and metastasis of HCC. Our findings provide miR-526b/SIRT7 axis as a promising drug target for HCC.

14.
Oncol Lett ; 12(4): 2648-2656, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27698837

ABSTRACT

Branched-chain amino acid transaminase 1 (BCAT1) has been associated with numerous types of tumors; however, few previous studies have evaluated the expression and role of BCAT1 in hepatocellular carcinoma (HCC). In the present study, the expression of BCAT1 was detected by reverse transcription-quantitative polymerase chain reaction and immunoblotting in six HCC cell lines and 74 pairs of HCC and adjacent non-cancerous liver tissues. In addition, the correlation between the expression levels of c-Myc and BCAT1 was analyzed using immunohistochemistry. Furthermore, RNA silencing was performed using c-Myc-specific or BCAT1-specific small interfering RNA, after which wound healing and Transwell cell invasion assays were performed. Finally, the clinicopathological characteristics of BCAT1 in patients with HCC were analyzed. It was shown that the expression of BCAT1 was significantly higher in HCC tissues compared with adjacent non-tumor tissues (P<0.001), and in HCC cell lines compared within the L-02 hepatic cell line (P<0.001). In addition, immunohistochemical analyses indicated that the expression of BCAT1 was positively correlated with c-Myc (r=0.706, P<0.001). BCAT1 expression was shown to be downregulated in c-Myc-knockdown cells, and silencing of BCAT1 expression reduced the invasion and migration of HCC cells. Furthermore, a clinical analysis indicated that BCAT1 expression in HCC tissues was significantly associated with the tumor-node-metastasis stage, tumor number and tumor differentiation (all P<0.05), and that BCAT1 was able to predict the 5-year survival and disease-free survival rates of patients with HCC (both P<0.001). The results of the present study suggested that BCAT1 expression is upregulated in patients with HCC, and that BCAT1 may serve as a potential molecular target for the diagnosis and treatment of HCC.

15.
Biomed Rep ; 5(4): 483-490, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27699018

ABSTRACT

A systematic review and meta-analysis were conducted to evaluate the efficacy and safety of early prophylactic anticoagulation for the prevention of portal vein system thrombosis (PVST) after splenectomy. A systematic search of the PubMed, EMBASE, Springer and Cochrane Library databases was performed to identify studies comparing the outcomes in patients receiving or not receiving regular prophylactic anticoagulation after splenectomy. The quality of the included studies was assessed using the Jadad Score and the Newcastle-Ottawa Scale. Heterogeneity was evaluated using the χ2 and I2 tests. The parameters that were analyzed included the incidence of PVST and anticoagulation-associated complications. A total of seven studies qualified for the review, involving 383 and 283 patients receiving or not receiving regular prophylactic anticoagulation, respectively. The incidence of PVST was significantly reduced with an odds ratio (OR) of 0.31 [95% confidence interval (CI), 0.21-0.46; P<0.00001] in the regular prophylactic anticoagulation group compared with the control group. No difference in the incidence of anticoagulation-associated complications was identified between the two groups (OR=0.60, 95% CI, 0.23-1.56; P=0.30). Early prophylactic anticoagulation was associated with a reduced incidence of PVST, although it was not associated with the incidence of anticoagulation-associated complications. These results indicate that prophylactic anticoagulation could be safely administered after splenectomy, even to cirrhotic patients.

16.
Cancer Lett ; 375(1): 190-198, 2016 May 28.
Article in English | MEDLINE | ID: mdl-26945969

ABSTRACT

UNLABELLED: The p300-CBP-associated factor (PCAF), other than its histone acetyltransferase (HAT) activity, possesses an intrinsic ubiquitination activity that is involved in various transcriptional regulators, including the transcription factor glioma-associated oncogene 1 (Gli1), a well-known regulator of epithelial-mesenchymal transition (EMT) in cancer. In present research, we detected that PCAF was down-regulated in hepatocellular carcinoma (HCC) tissues compared with the adjacent non-tumor tissues and significantly associated with malignant portal vein invasion (p < 0.05) and poor survival (p < 0.05) of HCC patients. Moreover, functional study demonstrated that downregulation of PCAF facilitated tumor cell migration, invasion via EMT. Further study found that Gli1 as a direct target of PCAF induced EMT and promoted tumor metastasis and invasion. CONCLUSION: PCAF is an anti-oncogene that plays an important role in the development of HCC by suppressing HCC cell metastasis and EMT by targeting Gli1, which indicates the potential therapeutic value of PCAF for suppression of metastasis of HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Epithelial-Mesenchymal Transition , Liver Neoplasms/metabolism , Lung Neoplasms/metabolism , Transcription Factors/metabolism , p300-CBP Transcription Factors/physiology , Animals , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Female , Humans , Kaplan-Meier Estimate , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Male , Mice, Nude , Middle Aged , Neoplasm Transplantation , Zinc Finger Protein GLI1
17.
BMC Cancer ; 16: 103, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26879601

ABSTRACT

BACKGROUND: The deregulation of E-cadherin has been considered as a leading cause of hepatocellular carcinoma (HCC) metastasis. BCL6 corepressor-like 1 (BCORL1) is a transcriptional corepressor and contributes to the repression of E-cadherin. However, the clinical significance of BCORL1 and its role in the metastasis of HCC remain unknown. METHODS: Differentially expressed BCORL1 between HCC and matched tumor-adjacent tissues, HCC cell lines and normal hepatic cell line were detected by Western blot. The expression of BCORL1 was altered by siRNAs or lentivirus-mediated vectors. Transwell assays were performed to determine HCC cell invasion and migration. RESULTS: Increased expression of BCORL1 protein was detected in HCC specimens and cell lines. Clinical association analysis showed that BCORL1 protein was expressed at significant higher levels in HCC patients with multiple tumor nodes, venous infiltration and advanced TNM tumor stage. Survival analysis indicated that high expression of BCORL1 protein conferred shorter overall survival (OS) and recurrence-free survival (RFS) of HCC patients. Multivariate Cox regression analysis disclosed that BCORL1 expression was an independent prognostic marker for predicting survival of HCC patients. Our in vitro studies demonstrated that BCORL1 prominently promoted HCC cell migration and invasion. Otherwise, an inverse correlation between BCORL1 and E-cadherin expression was observed in HCC tissues. BCORL1 inversely regulated E-cadherin abundance and subsequently facilitated epithelial-mesenchymal transition (EMT) in HCC cells. Notably, the effect of BCORL1 knockdown on HCC cells was abrogated by E-cadherin silencing. CONCLUSIONS: BCORL1 may be a novel prognostic factor and promotes cell migration and invasion through E-cadherin repression-induced EMT in HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Movement/genetics , Genetic Markers/genetics , Liver Neoplasms/metabolism , Neoplasm Invasiveness/genetics , Repressor Proteins/metabolism , Cadherins , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Female , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Male , Middle Aged , Prognosis , Repressor Proteins/analysis , Repressor Proteins/genetics
18.
Chin Med J (Engl) ; 128(22): 3043-9, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26608984

ABSTRACT

BACKGROUND: Intraperitoneal lymphangioma (IL) used to be thought of as a benign lymphatic malformation with a low rate of preoperative diagnosis. This retrospective study aimed to explore the connection between the cysts and clinical manifestation and imaging characteristics, and to study diagnostic confusion, therapeutic principles and potential recurrent reasons, to further enhance the comprehension of this rare disease. METHODS: Here, we retrospectively reviewed 21 patients diagnosed with IL. Age, sex, complaints, physical findings, and imaging features of each patient were documented. The therapies, postoperative complications and treatments were discussed. RESULTS: Symptomatology included eight patients (38%) with intermittent dull pain in the abdomen, and three patients (14%) complained of abdominal persistent pain. The physical examination revealed an abdominal mass in 16 patients (76%), and eight (38%) were reported no discomfort. IL was correctly established preoperatively in 19 patients (90%). Patients were treated using laparotomy, except one who was treated with laparoscopy. Two recurrences were noted during follow-up. CONCLUSIONS: IL should be suspected in any patient with a mobile abdominal mass and surgery is required immediately after discovery of the tumor.


Subject(s)
Lymphangioma/diagnosis , Abdomen/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Diagnosis, Differential , Female , Humans , Laparoscopy , Lymphangioma/surgery , Male , Middle Aged , Postoperative Complications , Retrospective Studies , Young Adult
19.
Mol Med Rep ; 12(4): 5997-6004, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26252226

ABSTRACT

High­mobility group box 1 (HMGB1), a member of the high­mobility group protein family, was originally characterized as a non­histone, nuclear DNA­binding protein. While the roles of HMGB1 in inflammation and cell differentiation have been previously reported, its role in tumor cell migration and invasion, particularly in hepatocellular carcinoma (HCC), has remained elusive. The present study reported that the expression of HMGB1 in HCC tissues was significantly higher than that in matched tumor­adjacent tissues (P<0.05). HMGB1 was expressed at significantly elevated levels in tumors of patients with large tumor size, high histological grade and advanced tumor­node­metastasis stage (P<0.05). The positive expression of HMGB1 correlated with a poor three­year overall and disease­free survival of HCC patients (P<0.05). In addition, HMGB1 was an independent factor for predicting the three­year overall and disease­free survival of HCC patients (P<0.05). An in vitro experiment revealed that knockdown of HMGB1 inhibited cell migration and invasion in the HCC cell lines Huh7 and MHCC97H (P<0.05). Furthermore, western blot analysis showed that HMGB1 knockdown markedly inhibited epithelial mesenchymal transition in Huh7 and MHCC97H cells. These results suggested that HMGB1 may be utilized as an independent prognostic marker in HCC and may promote tumor progression by promoting cell migration and invasion.


Subject(s)
Carcinoma, Hepatocellular/diagnosis , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , HMGB1 Protein/metabolism , Liver Neoplasms/diagnosis , Adult , Aged , Biomarkers, Tumor , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement , Disease-Free Survival , Female , HMGB1 Protein/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Middle Aged , Prognosis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transfection
20.
Oncotarget ; 6(15): 13216-28, 2015 May 30.
Article in English | MEDLINE | ID: mdl-25965836

ABSTRACT

MicroRNA-212 (miR-212) has been reported to play oncogenic or tumor suppressive role in different human malignancies. Here, we demonstrated that the mean level of miR-212 in hepatocellular carcinoma (HCC) tissues was significantly lower than that in matched tumor-adjacent tissues. Similarly, the expression of miR-212 was obviously reduced in HCC cell lines as compared with a nontransformed hepatic cell line. Ectopic expression of miR-212 inhibited cell viability and proliferation, and induced apoptosis in HepG2 cells. In contrast, down-regulation of miR-212 increased cell viability and proliferation, and suppressed apoptosis in Bel-7402 cells. In vivo studies showed that miR-212 inhibited tumor growth of HCC via suppressing proliferation and inducing apoptosis. Furthermore, we confirmed that Forkhead box protein A1 (FOXA1) was a direct target of miR-212, and it abrogated the function of miR-212 in HCC. Finally, we disclosed that the aberrant expression of miR-212 and FOXA1 was evidently correlated with poor prognostic features of HCC. MiR-212, FOXA1 and their combination were valuable prognostic markers for predicting survival of HCC patients. In conclusion, miR-212 may serve as a prognostic indicator for HCC patients and exerts tumor suppressive role, at least in part, by inhibiting FOXA1.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Hepatocyte Nuclear Factor 3-alpha/metabolism , Liver Neoplasms/drug therapy , MicroRNAs/pharmacology , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/physiology , Female , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...