Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
BMC Gastroenterol ; 24(1): 341, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39354355

ABSTRACT

BACKGROUND: Colonoscopic enteral tube placement using current methods has some shortcomings, such as the complexity of the procedure and tube dislodgement. The magnetic navigation technique (MNT) has been proven effective for nasoenteral feeding tube placement, and is associated with reduced cost and time to initiation of nutrition. This study attempted to develop a novel method for enteral tube placement using MNT. METHODS: The MNT device consisted of an external magnet and a 12 Fr tube with a magnet at the end. Ten swine were used, and bowel cleansing was routinely performed before colonoscopy. Intravenous anesthesia with propofol and ketamine was administered. A colonoscopic enteral tube was placed using the MNT. The position of the end of the enteral tube was determined by radiography, and angiography was performed to check for colonic perforations. Colonoscopy was used to detect intestinal mucosal damage after tube removal. RESULTS: MNT-assisted colonoscopic enteral tube placement was successfully completed in all pigs. The median operating time was 30 (26-47) min. No colon perforation was detected on colonography after enteral tube placement, and no colonic mucosal bleeding or injury was detected after the removal of the enteral tube. CONCLUSIONS: MNT-assisted colonoscopic enteral tube placement is feasible and safe in swine and may represent a valuable method for microbial therapy, colonic drainage, and host-microbiota interaction research in the future.


Subject(s)
Colonoscopy , Intubation, Gastrointestinal , Animals , Colonoscopy/methods , Swine , Intubation, Gastrointestinal/methods , Enteral Nutrition/methods , Enteral Nutrition/instrumentation , Magnets , Colon/diagnostic imaging , Feasibility Studies , Female , Operative Time
2.
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 inĀ 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
3.
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
4.
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
7.
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
8.
Cell Physiol Biochem ; 35(3): 983-96, 2015.
Article in English | MEDLINE | ID: mdl-25660117

ABSTRACT

BACKGROUND & AIMS: To investigate the expression and prognostic value of MACC1 in patients with HCC and identify the mechanism by which MACC1 inhibits HCC cell apoptosis. METHODS: MACC1 and p-AKT expression was studied using immunohistochemistry of both HCC tissues and adjacent liver tissues. qRT-PCR and western immunoblotting were used to examine the expression of target genes at the mRNA and protein levels, respectively. The MTT assay was used to assess cell viability, and cell apoptosis was determined by DAPI staining, Annexin V/PI staining and Caspase 3/7 assay. Nude mice were used to perform in vivo experiments. RESULTS: The overexpression of MACC1 was found in HCC tissues and was correlated with poor postsurgical prognosis. There was a positive relationship between MACC1 and p-AKT expression in HCC tissues. In vitro experiments showed that MACC1 repressed HCC cell apoptosis and promoted cell growth. Knockdown of c-MET abolished the anti-apoptotic function of MACC1. Next, MACC1 was verified to activate PI3K/AKT signaling by sensitizing HGF/c-MET signaling in HCC. MACC1 overexpression enhanced the HGF-driven phosphorylation of BAD, Caspase 9 and FKHRL1 and inhibited their pro-apoptotic functions in HCC cells. Finally, MACC1 was shown to inhibit cell apoptosis and promote HCC growth in vivo. CONCLUSIONS: This investigation revealed that MACC1 overexpression predicted worse prognosis after liver resection, which was attributed to the repression of HCC cell apoptosis via a molecular mechanism in which MACC1 accelerated the activation of the HGF/c-MET/PI3K/AKT pathway and phosphorylated BAD, Caspase 9 and FKHRL1, ultimately preventing their nuclear translocation and their pro-apoptotic function.


Subject(s)
Carcinoma, Hepatocellular/genetics , Hepatocyte Growth Factor/biosynthesis , Liver Neoplasms/genetics , Oncogene Protein v-akt/biosynthesis , Proto-Oncogene Proteins c-met/genetics , Transcription Factors/biosynthesis , Animals , Apoptosis , Carcinoma, Hepatocellular/pathology , Caspase 9/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/genetics , Heterografts , Humans , Liver Neoplasms/pathology , Mice , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/genetics , RNA, Messenger , Signal Transduction , Trans-Activators , Transcription Factors/genetics , bcl-Associated Death Protein/metabolism
9.
Hepatol Res ; 45(8): 906-18, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25263743

ABSTRACT

AIM: Targeting protein for Xenopus kinesin-like protein 2 (TPX2) is a microtubule-associated protein that impacts spindle assembly in human cells. Several studies have shown that the overexpression of TPX2 is correlated with multiple tumor types. However, the role of TPX2 in hepatocellular carcinoma (HCC) remains undetermined. METHODS: TPX2 expression was detected by quantitative reverse transcription polymerase chain reaction and immunoblotting in six cell lines and 130 pairs of HCC and adjacent non-cancerous liver tissues. Matrix metalloproteinase (MMP)2 and MMP9 expression was detected by immunohistochemistry to evaluate their correlations with TXP2. TPX2 siRNA was used to knock down TPX2 expression, and cell migration and invasion were determined. Moreover, clinical significance of TPX2 in HCC was analyzed. RESULTS: TPX2 expression was found to be obviously higher in HCC tissues than that in non-tumor tissues. Elevated TPX2 expression was observed in HCC cell lines as compared with that in a non-transformed hepatic cell line. Clinical analysis indicated that TPX2 expression in the HCC tissues was evidently correlated with the tumor-node-metastasis stage, tumor number and tumor differentiation. TPX2 is a novel prognostic marker for predicting 5-year overall survival and disease-free survival of HCC patients. TPX2 expression was positively correlated with MMP2 and MMP9. In vitro studies found that TPX2 knockdown prominently reduced cell invasion and migration and decreased phosphorylated AKT, MMP2 and MMP9 expression in MHCC-97H cells. CONCLUSION: Overexpression of TPX2 was associated with clinicopathological characteristics and poor patient outcomes. TPX2 may serve as a novel prognostic marker for HCC.

10.
Mol Cancer ; 13: 110, 2014 May 17.
Article in English | MEDLINE | ID: mdl-24884509

ABSTRACT

BACKGROUND: The E3 ubiquitin ligase Fbxw7 functions as a general tumor suppressor by targeting several well-known oncoproteins for ubiquitination and proteasomal degradation. However, the clinical significance of Fbxw7 and the mechanisms involved in the anti-cancer effect of Fbxw7 in HCC are not clear. METHOD: The Fbxw7 and YAP expression in 60 samples of surgical resected HCC and matched normal tumor-adjacent tissues were assessed using IHC or immunoblotting. Flow cytometry, caspase 3/7 activity assay, BrdU cell proliferation assay and MTT assay were used to detect proliferation and apoptosis of HCC cells. The regulatory effect of Fbxw7 on YAP in HCC cells was confirmed by qRT-PCR, immunoblotting and immunofluorescence. Co-immunoprecipitation was used to analyze interaction between YAP and Fbxw7. Nude mice subcutaneous injection, Ki-67 staining and TUNEL assay were used to evaluate tumor growth and apoptosis in vivo. RESULTS: In this study, we found that Fbxw7 expression was impaired in HCC tissues and loss of Fbxw7 expression was correlated with poor clinicopathological features including large tumor size, venous infiltration, high pathological grading and advanced TNM stage. Additionally, we demonstrated that patients with positive Fbxw7 expression had a better 5-year survival and Fbxw7 was an independent factor for predicting the prognosis of HCC patients. We confirmed that Fbxw7 inhibited HCC by inducing both apoptosis and growth arrest. Elevated YAP expression was observed in the same cohort of HCC tissues. Pearson's correlation coefficient analysis indicated that Fbxw7 was inversely associated with YAP protein expression in HCC tissues. We also found that Fbxw7 regulated YAP protein abundance by targeting YAP for ubiquitination and proteasomal degradation in HCC. Furthermore, restoring YAP expression partially abrogated Fbxw7 induced HCC cell apoptosis and growth arrest in vitro and in vivo. CONCLUSION: These results indicate that Fbxw7 may serve as a prognostic marker and that YAP may be a potential target of Fbxw7 in HCC.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Cell Cycle Proteins/genetics , F-Box Proteins/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Phosphoproteins/genetics , Ubiquitin-Protein Ligases/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Female , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Grading , Neoplasm Staging , Neoplasm Transplantation , Phosphoproteins/metabolism , Prognosis , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Signal Transduction , Transcription Factors , Tumor Microenvironment , Ubiquitin-Protein Ligases/metabolism , YAP-Signaling Proteins
11.
BMC Cancer ; 14: 938, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25494879

ABSTRACT

BACKGROUND: Fibulin-5 has been considered as a tumor suppressor through inhibiting tumor growth and invasion. Reduced expression of Fibulin-5 is frequently observed in various human cancers. In this study, we investigate the clinical significance of Fibulin-5 and its role in hepatocellular carcinoma (HCC) cell migration and invasion. METHODS: The expression of Fibulin-5 was evaluated by qRT-PCR and immunoblotting in HCC and matched noncancerous tissues. Fibulin-5 was over-expressed or knocked down by a retrovirus-mediated expression plasmid or a specific siRNA in HCC cells. Boyden chamber and Transwell assays were used to test HCC cell migration and invasion. Immunostaining was performed to determine matrix metalloproteinase-7 (MMP-7) expression in HCC specimens. MMP-7 retroviruses and siRNA were used to alter MMP-7 expression in HCC cells. RESULTS: In our study, the expression levels of Fibulin-5 protein and mRNA were down-regulated in HCC tissues as compared with those in matched noncancerous tissues. Reduced expression of Fibulin-5 was observed in all HCC cell lines (HepG2, SMMC-7721, MHCC97L, Hep3B, MHCC97H and HCC-LM3) as compare with that in a non-transformed hepatic cell line (LO2). Low expression of Fibulin-5 was significantly correlated with poor prognostic features including multiple tumor nodes, venous infiltration, high Edmondson-Steiner grading and advanced tumor-node-metastasis (TNM) tumor stage. Furthermore, we demonstrated that Fibulin-5 was a novel independent prognostic marker for predicting 5-year survival of HCC patients. Our in vitro studies showed that Fibulin-5 overexpression inhibited HCC cell migration and invasion. While Fibulin-5 knockdown increased the number of migrated and invaded HCC cells. Fibulin-5 negatively regulated MMP-7 abundance in HCC cells. Moreover, the inverse correlation between Fibulin-5 and MMP-7 expressions was observed in HCC tissues. Mechanistically, we disclosed that MMP-7 knockdown reduced the number of migrated and invaded HCC cells. Restoring MMP-7 expression abrogated the suppressive effect of Fibulin-5 on HCC cell migration and invasion in vitro, suggesting that Fibulin-5 exerted its anti-metastatic function, at least in part, by down-regulating the expression of MMP-7 in HCC cells. CONCLUSIONS: These results indicate that Fibulin-5 may serve as a prognostic biomarker and inhibits HCC invasion and metastasis by suppressing MMP-7 expression.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Extracellular Matrix Proteins/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Matrix Metalloproteinase 7/metabolism , RNA, Messenger/analysis , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/chemistry , Cell Migration Assays , Cell Movement , Down-Regulation , Extracellular Matrix Proteins/analysis , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hep G2 Cells , Humans , Liver/chemistry , Liver Neoplasms/chemistry , Liver Neoplasms/secondary , Lymphatic Irradiation , Male , Matrix Metalloproteinase 7/analysis , Matrix Metalloproteinase 7/genetics , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Survival Rate , Tumor Burden
12.
Int J Mol Sci ; 15(11): 20486-99, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25387077

ABSTRACT

MircroRNA-130b (miR-130b) is proposed as a novel tumor-related miRNA and has been found to be significantly dysregulated in tumors. In this study, the expression level of miR-130b was found to be obviously higher in hepatocellular carcinoma (HCC) tissues than that in nontumor tissues. Further, miR-130b was expressed at significantly higher levels in aggressive and recurrent tumor tissues. Clinical analysis indicated that high-expression of miR-130b was prominently correlated with venous infiltration, high Edmondson-Steiner grading and advanced tumor-node-metastasis (TNM) tumor stage in HCC. Elevated miR-130b expression was observed in all HCC cell lines (HepG2, SMMC-7721, Huh7, Hep3B and MHCC97H) as compared with that in a nontransformed hepatic cell line (LO2). Furthermore, an inverse correlation between miR-130b and E-cadherin and a positive correlation between miR-130b and Vimentin were observed in HCC tissues. Down-regulation of miR-130b expression reduced invasion and migration in both Hep3B and MHCC97H cells. Peroxisome proliferator-activated receptor gamma (PPAR-ƎĀ³) was inversely correlated with miR-130b expression in HCC tissues. In addition, down-regulation of miR-130b restored PPAR-ƎĀ³ expression and subsequently suppressed epithelial-mesenchymal transition (EMT) in HCC cells. We identified PPARƎĀ³ as a direct target of miR-130b in HCC in vitro. Notably, PPAR-ƎĀ³ knockdown abolished down-regulation of miR-130b-inhibited EMT in MHCC97H cells. In conclusion, miR-130b may promote HCC cell migration and invasion by inhibiting PPAR-ƎĀ³ and subsequently inducing EMT.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Liver/pathology , MicroRNAs/metabolism , PPAR gamma/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Liver/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , MicroRNAs/analysis , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , PPAR gamma/analysis , PPAR gamma/genetics
13.
Int J Mol Sci ; 15(5): 7124-38, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24776759

ABSTRACT

Sterol regulatory element-binding protein 1 (SREBP-1) is a well-known nuclear transcription factor involved in lipid synthesis. Recent studies have focused on its functions in tumor cell proliferation and apoptosis, but its role in cell migration and invasion, especially in hepatocellular carcinoma (HCC), is still unclear. In this study, we found that the expression of SREBP-1 in HCC tissues was significantly higher than those in matched tumor-adjacent tissues (p < 0.05). SREBP-1 was expressed at significantly higher levels in patients with large tumor size, high histological grade and advanced tumor-node-metastasis (TNM) stage (p < 0.05). The positive expression of SREBP-1 correlated with a worse 3-year overall and disease-free survival of HCC patients (p < 0.05). Additionally, SREBP-1 was an independent factor for predicting both 3-year overall and disease-free survival of HCC patients (p < 0.05). In vitro studies revealed that downregulation of SREBP-1 inhibited cell proliferation and induced apoptosis in both HepG2 and MHCC97L cells (p < 0.05). Furthermore, wound healing and transwell assays showed that SREBP-1 knockdown prominently inhibited cell migration and invasion in both HepG2 and MHCC97L cells (p < 0.05). These results suggest that SREBP-1 may serve as a prognostic marker in HCC and may promote tumor progression by promoting cell growth and metastasis.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Liver/pathology , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Sterol Regulatory Element Binding Protein 1/analysis , Adult , Aged , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver/metabolism , Liver Neoplasms/diagnosis , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Middle Aged , Neoplasm Invasiveness/diagnosis , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/diagnosis , Neoplasm Metastasis/genetics , Prognosis , RNA Interference , RNA, Small Interfering/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Up-Regulation
14.
Chin J Cancer Res ; 26(4): 410-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25232213

ABSTRACT

OBJECTIVE: Targeting protein for Xenopus kinesin-like protein 2 (TPX2) is a nuclear proliferation-related protein that plays a critical role in the formation of mitotic spindle. High expression of TPX2 has been observed in several types of tumors. However, the role of TPX2 in hepatocellular carcinoma (HCC) remains unclear. Our study aimed to investigate the effect of TPX2 on HCC cell invasion. METHODS: The immortalized normal human liver cell line L02 and six HCC cell lines including SMMC-7721, BEL-7402, Huh-7, HepG2, Hep3B and SKHep1 were subjected to qRT-PCR and western blot for TPX2 mRNA and protein, respectively. Furthermore, TPX2 small interfering RNA (siRNA) was used to knock down TPX2 expression in SMMC-7721 and HepG2 cells. Cell proliferation and invasion were determined by MTT and transwell assays. Otherwise, expression of p-AKT, MMP2 and MMP9 were evaluated by western blot in SMMC-7721 cells. RESULTS: The expression of TPX2 in HCC cell lines was markedly higher than that in normal human liver cell line. TPX2 knockdown using a specific TPX2-siRNA reduced the number of invaded cells and inhibited cell proliferation in SMMC-7721 and HepG2 cells. Furthermore, TPX2 knockdown resulted in inactivation of AKT signaling and down-regulation of MMP2 and MMP9 expression in SMMC-7721 cells. CONCLUSIONS: Our study identified that TPX2 might contribute to tumor cell invasion through activating AKT signaling and subsequently increasing MMP2 and MMP9 in HCC.

15.
Mater Today Bio ; 28: 101169, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39183770

ABSTRACT

Thanks to its structural characteristics and signal patterns similar to those of human brain synapses, memristors are widely believed to be applicable for neuromorphic computing. However, to our knowledge, memristors have not been effectively applied in the biomedical field, especially in disease diagnosis and health monitoring. In this work, a blood-based biomemristor was prepared for in vitro detection of hyperglycemia and hyperlipidemia. It was found that the device exhibits excellent resistance switching (RS) behavior at lower voltage biases. Through mechanism analysis, it has been confirmed that the RS behavior is driven by Ohmic conduction and ion rearrangement. Furthermore, the hyperglycemia and hyperlipidemia detection devices were constructed for the first time based on memristor logic circuits, and circuit simulations were conducted. These results confirm the feasibility of blood-based biomemristors in detecting hyperglycemia and hyperlipidemia, providing new prospects for the important application of memristors in the biomedical field.

16.
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 1000Ā 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.

17.
Mol Cancer ; 12(1): 96, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23981651

ABSTRACT

BACKGROUND: PCAF is an important intrinsic histone acetyltransferases. This study tried to establish the effect of PCAF on HCC cell apoptosis. METHOD: Both in vitro and in vivo experiments including IHC, DAPI staining, caspase 3/7 activity assay, BrdU assay, MTT assay, western immunoblotting and co-immunoprecipitation were used here. RESULTS: PCAF was found to be expressed at the low level in most of HCC cell lines. PCAF overexpression induced cell apoptosis and growth arrest with increased Histone H4 acetylation and inactivation of AKT signaling in Huh7 and HepG2 cells. The opposite results were obtained by silencing PCAF in Hep3B cells. The co-immunoprecipitation assay confirmed that PCAF protein was bound with histone H4 protein in the nucleus of Hep3B cells. Finally, the in vivo experiment confirmed the findings mentioned-above. CONCLUSION: These data identified PCAF promotes cell apoptosis and functions as a HCC repressor through acetylating histone H4 and inactivating AKT signaling.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/enzymology , Histones/metabolism , Liver Neoplasms, Experimental/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , p300-CBP Transcription Factors/physiology , Acetylation , Animals , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Gene Knockdown Techniques , Hep G2 Cells , Humans , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation , Signal Transduction , Tumor Burden , Up-Regulation
18.
Mol Cell Biochem ; 376(1-2): 21-32, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23232575

ABSTRACT

Expression of MACC1 (metastasis-associated in colon cancer-1) protein is associated with metastasis of various human cancers. This study analyzed MACC1 protein expression in hepatocellular carcinoma (HCC) tissue specimens and then investigated the effects of MACC1 knockdown on HCC cell migration and invasion, and gene expression levels. Sixty pairs of HCC and adjacent normal liver tissues from HCC patients were analyzed for MACC1 expression immunohistochemically. The HCC cell lines Hep3B, Huh7, MHCC97H, SMMC-7721, Bel-7402, and HepG2 and the normal liver cell line LO2 were used to assess expressions of MACC1 mRNA and MACC1 protein using qRT-PCR and western blot, respectively. MACC1 short hairpin RNA (shRNA) was used to knockdown MACC1 protein expression in Huh7 cells. Changes in the tumor phenotype of these cells were analyzed with wound healing assay and invasion assays, and differences in gene expression were evaluated via western blot. Immunofluorescence was used to locate MACC1 protein in the above cell lines. MACC1 was highly expressed in HCC tissues and the nuclear expression of MACC1 protein was associated with poor tumor differentiation and intrahepatic metastasis or portal invasion. Moreover, MACC1 mRNA and MACC1 protein was also expressed in HCC cell lines. Immunostaining showed that MACC1 protein was localized in both nuclei and cytoplasm of HCC cell lines and the nuclear localization of MACC1 protein was associated with increased aggressiveness of HCC in cell lines. Knockdown of MACC1 expression using MACC1-shRNA reduced Huh7 cell migration and invasion abilities, which was associated with downregulation of MMP2, MMP9, and c-Met proteins in Huh7 cells. Localization of MACC1 protein to the nucleus may predict HCC progression. Knockdown of MACC1 expression using MACC1 shRNA warrants further evaluation as a novel therapeutic strategy for control of HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Transcription Factors/genetics , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Nucleus/metabolism , Cytosol/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hep G2 Cells , Humans , Liver/metabolism , Liver Neoplasms/genetics , Proto-Oncogene Proteins c-met/metabolism , RNA, Small Interfering , Reference Values , Trans-Activators , Transcription Factors/metabolism
19.
Surg Innov ; 20(1): 32-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22461108

ABSTRACT

Open splenectomy and esophagogastric devascularization (OSED) is a typical surgery for portal hypertension. Because of the high morbidity associated with it, it is desirable to develop a minimally invasive alternative. To investigate the safety and effect of laparoscopic splenectomy and esophagogastric devascularization (LSED), we performed LSED for 24 patients suffering from portal hypertension with refractory variceal bleeding while conducting OSED for 30 patients. The perioperative data and follow-up results were analyzed. Operation times were similar in both groups. Less intraoperative blood and faster return of gastrointestinal function were found in the LSED group. The LSED group had lower levels of alanine aminotransferase, aspartate aminotransferase, and total bilirubin after surgery. In both groups, the levels of platelet count, white blood cell count, or hemoglobin were increased after operation dramatically. During the follow-up period (range = 3-36 months), no patient had recurrent hypersplenism or variceal bleeding. Hence, LSED is a safe and minimally invasive intervention for portal hypertension with refractory variceal bleeding.


Subject(s)
Esophagoscopy/methods , Gastrointestinal Hemorrhage/surgery , Hypertension, Portal/surgery , Laparoscopy/methods , Minimally Invasive Surgical Procedures/methods , Splenectomy/methods , Adolescent , Adult , Aged , Alanine Transaminase/analysis , Esophagoscopy/adverse effects , Esophagoscopy/statistics & numerical data , Female , Gastrointestinal Hemorrhage/etiology , Humans , Hypertension, Portal/complications , Intraoperative Period , Laparoscopy/adverse effects , Laparoscopy/statistics & numerical data , Liver Function Tests , Male , Middle Aged , Minimally Invasive Surgical Procedures/adverse effects , Minimally Invasive Surgical Procedures/statistics & numerical data , Retrospective Studies , Splenectomy/adverse effects , Splenectomy/statistics & numerical data , Statistics, Nonparametric
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL