Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
2.
Int J Gen Med ; 15: 1677-1687, 2022.
Article in English | MEDLINE | ID: mdl-35210837

ABSTRACT

PURPOSE: Colon cancer is the most commonly diagnosed gastrointestinal cancer. This research intended to evaluate the prognostic values of LINC01006 and miR-3199 for colon cancer and their effects on cell physiology. PATIENTS AND METHODS: LINC01006 and miR-3199 expression levels were determined by RT-qPCR. Patients' 5-year cumulative survival rate was analyzed by Kaplan-Meier curves with the Log rank test. Chi-square test and multivariate Cox regression analysis were used to access the clinical significance. CCK-8 assay, transwell assay, and TUNEL assays were used to monitor the change of cell proliferation, invasion, migration, and apoptosis. RESULTS: The expression level of LINC01006 was increased while miR-3199 was decreased in colon tissues and cells compared to normal ones. This dysregulated expression was correlated with T stage (P = 0.002) and N stage (P = 0.009). High LINC01006 level (HR = 4.048, 95%: 1.502-10.911, P = 0.006) or low miR-3199 level (HR = 3.421, 95% CI: 1.254-9.330, P = 0.016) was outstanding for predicting poor prognosis in patients with colon cancer. Downregulation of LINC01006 reduced cell proliferation, invasion, and migration but induced cell apoptosis (P < 0.05). CONCLUSION: LINC01006 knockdown showed anti-proliferative, anti-metastatic, and apoptotic-induced effects on colon cancer cells. This study contributes to research on promising prognostic biomarkers of colon cancer and might give way to further investigation of alternative tumor targets.

3.
Asian J Surg ; 45(1): 332-338, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34147329

ABSTRACT

BACKGROUND: Current management guidelines for gallbladder polyps (GBPs) focus on a diameter more than 1 cm as an indication for cholecystectomy. Since most GBPs are not malignant, unnecessary cholecystectomies can lead to unnecessary complications and costs. We developed a score to identify true polyps focusing on their cross-sectional area (CSA). METHODS: We retrospectively analyzed the demographic, clinical, laboratory, and sonographic characteristics of 522 patients with GBPs who had undergone cholecystectomy at our hospital between January 2010 and July 2020 (reference group). We used univariate analysis to compare these parameters between 88 true polyps and 434 pseudopolyps and multivariate logistic regression analysis to identify parameters to include in our scoring model. Receiver operating characteristics and area under the curve were used to identify cut-off values. The model was tested on a validation group of 98 patients. RESULTS: In the multivariate analysis, a CSA >123 mm2, positive blood flow signal, age >55.5 years, alanine aminotransferase (ALT) levels > 50 U/L, and an ALT/aspartate aminotransferase ratio > 0.77 were significantly associated with true polyps (odds ratio 6.528, 2.377, 2.617, 2.445, and -0.372, respectively). A prediction model based on cut-off values was used to distinguish a low-risk and high-risk GBP group; true polyps accounted for 6.54% and 58.72%, respectively (p < 0.001). In the low-risk and high-risk validation groups, true polyps comprised 12.35% and 82.35%, respectively (p < 0.001). CONCLUSIONS: Our scoring system shows high accuracy and specificity in identifying true polyps and helps determine the need for surgical resection.


Subject(s)
Gallbladder Diseases , Gallbladder Neoplasms , Polyps , Gallbladder Diseases/diagnostic imaging , Gallbladder Diseases/surgery , Humans , Middle Aged , Polyps/diagnostic imaging , Polyps/surgery , Retrospective Studies
4.
J Exp Clin Cancer Res ; 40(1): 218, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34193219

ABSTRACT

BACKGROUND: Liver cancer stem cells (LCSCs) play key roles in the metastasis, recurrence, and chemotherapeutic resistance of hepatocellular carcinoma (HCC). Our previous research showed that the POSTN gene is closely related to the malignant progression and poor prognosis of HCC. This study aimed to elucidate the role of POSTN in generating LCSCs and maintaining their stemness as well as the underlying mechanisms. METHODS: Human HCC tissues and matched adjacent normal tissues were obtained from 110 patients. Immunohistochemistry, western blotting (WB), and RT-PCR were performed to detect the expression of POSTN and stemness factors. The roles of transforming growth factor (TGF)-ß1 and AP-2α in the POSTN-induced stemness transformation of HCC cells were explored in vitro and in vivo using LCSCs obtained by CD133+ cell sorting. RESULTS: The high expression of POSTN was correlated with the expression of various stemness factors, particularly CD133, in our HCC patient cohort and in TCGA and ICGC datasets. Knockdown of POSTN expression decreased the abilities of HCC cell lines to form tumours in xenograft mouse models. Knockdown of POSTN expression also suppressed cell viability and clone formation, invasion, and sphere formation abilities in vitro. Knockdown of AP-2α attenuated the generation of CD133+ LCSCs and their malignant behaviours, indicating that AP-2α was a critical factor that mediated the POSTN-induced stemness transformation and maintenance of HCC cells. The role of AP-2α was verified by using a specific αvß3 antagonist, cilengitide, in vitro and in vivo. Activation of POSTN could release TGFß1 from the extracellular matrix and initiated POSTN/TGFß1 positive feedback signalling. Furthermore, we found that the combined use of cilengitide and lenvatinib suppressed the growth of HCC cells with high POSTN expression more effectively than the use of lenvatinib alone in the patient-derived xenograft (PDX) mouse model. CONCLUSIONS: The POSTN/TGFß1 positive feedback pathway regulates the expression of stemness factors and the malignant progression of HCC cells by regulating the transcriptional activation of AP-2α. This pathway may serve as a new target for targeted gene therapy in HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Adhesion Molecules/metabolism , Liver Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Transcription Factor AP-2/metabolism , Transforming Growth Factor beta1/metabolism , Adult , Animals , Carcinoma, Hepatocellular/pathology , Cell Proliferation/physiology , Disease Models, Animal , Feedback, Physiological , Heterografts , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplastic Stem Cells/pathology
5.
Transl Cancer Res ; 10(3): 1578-1582, 2021 Mar.
Article in English | MEDLINE | ID: mdl-35116482

ABSTRACT

Cancer is a major worldwide public health problem. With the popularity of medical examinations, the improvement of surgical procedures and the application of antitumor drugs, the mortality rate of cancer has declined in recent years, but the quantity remains extremely high. At the same time, with the emergence of tumor drug resistance, it is particularly urgent to seek new and effective drugs for tumor treatment. Solanine, a type of steroidal alkaloid, is the main extract of Solanum nigrum L. Because of its wide biological activity, it has attracted increasing attention, specifically, towards the empirical study of its antitumor activity. This article summarizes the research progress of the antitumor effect and mechanism of solanine over the past 20 years, to provide a reference for workers in the field of cancer research. Studies from 2000 to 2020 were reviewed from PubMed, Springer Link and the Web of Science using the keywords Solanum nigrum L., solanine, α-solanine, ß-solanine, γ-solanine, tumor, cancer and their combinations. Exclude research articles of extraction mixture. Language is limited to English. Solanine shows antitumor ability against different tumors by targeting different proteins. Although there is a lack of clinical large-scale studies, a large number of basic pharmacological and toxicological studies have confirmed that solanine may be a new effective cancer drug or adjuvant therapy. Solanine preparations and solanine derivative preparations have broad economic and scientific research prospects.

7.
Immunopharmacol Immunotoxicol ; 42(6): 556-563, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32811220

ABSTRACT

BACKGROUND: Capsaicin is a chili pepper extract with multiple therapeutic properties including anti-liver fibrosis. However, the paucity of its underlying mechanisms limited its widely clinical application. METHODS: In the present study, carbon tetrachloride (CCl4) was used to induce liver fibrosis in mice, and transforming growth factorß1 (TGFß1) was used to mimic liver fibrosis in vitro. Flow cytometry was conducted to determine the expression of CD80. The inflammatory factors level was examined by ELISA, and gene expression was detected by real-time PCR and western blot. RESULTS: Here, we show that capsaicin attenuates liver fibrosis progression by mediating macrophage inflammatory response. Capsaicin inhibited M1 polarization of macrophage by regulating Notch signaling leading to the reduced secretion of inflammatory cytokine TNF-α that correspondingly attenuates myofibroblasts regeneration and fibrosis formation of hepatocyte stellate cells (HSCs). CONCLUSION: Taken together, capsaicin alleviates liver fibrosis by inactivation of Notch signaling and further inhibiting TNF-α secretion from M1 macrophage. Targeting TNF-α or Notch signaling in macrophage represents a promising strategy to combat liver fibrosis.


Subject(s)
Capsaicin/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Liver Cirrhosis, Experimental/prevention & control , Liver/drug effects , Macrophages/drug effects , Receptors, Notch/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Carbon Tetrachloride , Cell Line , Cell Proliferation/drug effects , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Macrophage Activation/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phenotype , Signal Transduction
8.
J BUON ; 25(2): 899-906, 2020.
Article in English | MEDLINE | ID: mdl-32521884

ABSTRACT

PURPOSE: The purpose of this study was to compare the long-term outcomes of laparoscopic and open sphincter-preserving total mesorectal excision (TME) for low rectal cancer (LRC) using propensity score matching (PSM). METHODS: The clinical and follow-up data of 169 patients with LRC who underwent sphincter-preserving TME at our institution between January 2011 and January 2014 were retrospectively analyzed. Patients were divided into laparoscopic and open group based on the surgical approach. PSM including age, sex, body mass index, clinical stage, and American Society of Anesthesiologists score with a 1:1 ratio was subsequently performed. Sixty-eight patients in each group were ultimately included, and short- and long-term outcomes were compared between groups. RESULTS: Compared with the open group, the laparoscopic group had less intraoperative blood loss, more rapid postoperative recovery, and lower incidence of 30-day postoperative complications. However, there were no significant differences in severity of postoperative 30-day complications between the two groups. Both groups had no intraoperative or 30-day postoperative mortality. Regarding survival outcome, tumor recurrence rate, tumor recurrence site, 5-year overall survival, and 5-year disease-free survival, there were no significant differences between groups. CONCLUSION: Laparoscopic sphincter-preserving TME can achieve long-term outcomes similar to those of open TME for LRC.


Subject(s)
Laparoscopy/methods , Minimally Invasive Surgical Procedures/methods , Rectal Neoplasms/surgery , Aged , Female , Humans , Male , Middle Aged , Propensity Score , Rectal Neoplasms/pathology , Retrospective Studies , Treatment Outcome
9.
J Biochem ; 167(5): 525-534, 2020 May 01.
Article in English | MEDLINE | ID: mdl-32044992

ABSTRACT

The apoptosis of hepatocytes contributes to the activation of hepatic stellate cells (HSCs), thus promoting the accumulation of extracellular matrix proteins and aggravating liver fibrosis. Silent information regulator 1 (SIRT1) is an anti-fibrotic protein whose downregulation induces hepatocyte apoptosis. This study aims to identify whether SIRT1 is regulated by long non-coding RNA LINC01093 and explore its underlying mechanisms. Liver fibrosis was induced in mice using CCl4, and the differential expressions of several fibrosis-related long noncoding RNAs were detected in liver tissues. The effect of LINC01093 on cell apoptosis and viability of hepatocytes were investigated after LINC01093 overexpression or knockdown using flow cytometry and MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay. The anti-fibrotic effect of LINC01093 overexpression was observed in vivo. LncRNA LINC01093 is downregulated in CCl4-induced liver tissues and TGF-ß1-stimulated hepatocytes. Downregulated LINC01093 promoted cell apoptosis and inhibited cell viability of hepatocytes. The co-culture between LINC01093-knockdown hepatocytes and HSCs increased the expressions of pro-fibrotic proteins. Downregulated LINC01093 promoted hepatocyte apoptosis via promoting degradation and ubiquitination of SIRT1 under TGF-ß1 stimulation. The injection of LINC01093-overexpressing vectors alleviated liver fibrosis in vivo. In liver fibrosis, the downregulated LINC01093 promoted hepatocyte apoptosis, which is mediated by increasing the degradation and ubiquitination of SIRT1.


Subject(s)
Apoptosis , Down-Regulation , Hepatocytes/metabolism , Liver Cirrhosis/metabolism , RNA, Long Noncoding/metabolism , Sirtuin 1/metabolism , Ubiquitination , Animals , Carbon Tetrachloride/administration & dosage , Cells, Cultured , Disease Models, Animal , Injections, Intraperitoneal , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred BALB C , RNA, Long Noncoding/genetics
10.
Exp Ther Med ; 17(1): 639-648, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30651845

ABSTRACT

Various microRNAs (miRNA) have been recognized potential novel tumor markers and have a critical role in cancer development and progression. Recently, methylation of miRNA-148a was identified as a crucial biochemical process in the progression of cancer. However, its potential role and in pancreatic cancer as well as the underlying mechanisms have remained largely elusive. The present study investigated the potential antitumor effect of miR-148a as well as its impact on invasion and metastasis in pancreatic cancer. It was found that the expression of miRNA-148a and the potential predictive biomarker maternally expressed gene-3 (MEG-3) were obviously decreased in human pancreatic cancer tissues compared with those in adjacent non-tumorous tissues. Furthermore, miR-148a was found to be downregulated in pancreatic cancer cell lines compared with normal pancreatic cells through promoter methylation. An MTT assay and a clonogenic assay demonstrated that restoration of miRNA-148a inhibited the proliferation and colony formation of pancreatic cancer cells. In addition, miR-148a transduction led to the upregulation of MEG-3 expression and promoted apoptosis of pancreatic cancer cells. Western blot analysis revealed that transduction of miR-148a markedly decreased the expression levels of C-myc, cyclin D1 and ß-catenin in pancreatic cancer cells. Methylation of miR-148a not only decreased the endogenous ß-catenin levels but also inhibited the nuclear translocation of ß-catenin to delay cell cycle progression. Furthermore, ectopic miR-148a methylation inhibited pancreatic cancer cell migration and invasion via causing an upregulation of MEG-3 expression. Most importantly, ectopic overexpression of miR-148a in pancreatic cancer cells inhibited tumor formation in an animal experiment. Taken together, miR-148a methylation is a crucial regulatory process to inhibit the proliferation and invasion of pancreatic cancer cells, and transduction of miR-148a suppressed the proliferation of pancreatic cancer cells through negative regulation of the Wnt/ß-catenin signaling pathway. The findings of the present study suggested that miRNA-148a acts as a tumor suppressor in pancreatic cancer and may contribute to the development of novel treatments for pancreatic cancer.

11.
Dig Dis Sci ; 64(6): 1560-1570, 2019 06.
Article in English | MEDLINE | ID: mdl-30673982

ABSTRACT

BACKGROUND: The activation of hepatic stellate cells (HSCs) is involved in hepatic fibrogenesis and is regulated by the decreased expression of peroxisome proliferator-activated receptor γ (PPARγ). Rosiglitazone (RGZ) is a highly potent agonist of PPARγ. AIMS: To clarify molecular regulatory mechanism of RGZ in the activation of HSCs in hepatic fibrosis. METHODS: A mouse model of hepatic fibrosis was established by carbon tetrachloride with or without RGZ intervention. A vector carrying pcDNA-HOTAIR was constructed and injected into a mouse model. HSCs were isolated from liver tissue and activated by transforming growth factor-ß. The expression of miR-124-3p, HOTAIR, Col1A1, α-SMA, and PPARγ mRNAs was measured by quantitative real-time PCR. The level of PPARγ was measured by Western blotting. The interaction between HOTAIR and PPARγ was assessed by RNA immunoprecipitation (RIP) and RNA pull-down. The target gene of miR-124-3p was determined by luciferase reporter assay and RNA interference approaches. RESULTS: The expression of Col1A1 and α-SMA was reduced after RGZ intervention. Different expressions of HOTAIR and miR-124-3p were observed in liver tissue and HSCs. The luciferase reporter assay and RNA interference approaches indicated that miR-124-3p negatively regulated HOTAIR expression. RIP and RNA pull-down results revealed that PPARγ was interacted by HOTAIR. The therapeutic effect of RGZ on hepatic fibrosis was reversed by overexpression of HOTAIR. CONCLUSIONS: RGZ inhibits the activation of HSCs by up-regulating miR-124-3p. The silencing of HOTAIR by miR-124-3p in HSC activation provided the foundation to understand interactions of ncRNAs and potential treatment target in hepatic fibrosis.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Hepatic Stellate Cells/drug effects , Liver Cirrhosis, Experimental/prevention & control , Liver/drug effects , MicroRNAs/metabolism , Rosiglitazone/pharmacology , Animals , Carbon Tetrachloride , Cells, Cultured , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Male , Mice, Inbred BALB C , MicroRNAs/genetics , PPAR gamma/agonists , PPAR gamma/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Signal Transduction/drug effects , Up-Regulation
12.
Oncogene ; 37(9): 1220-1236, 2018 03.
Article in English | MEDLINE | ID: mdl-29242604

ABSTRACT

Ribosomal protein s15a (RPS15A) plays a promotive role in the mRNA/ribosome interactions during early translation. Our previous study has found that inhibiting RPS15A expression can decrease proliferation and induce cell cycle arrest in hepatocellular carcinoma (HCC) cell lines. However, the mechanism underlying the involvement of RPS15A in HCC pathogenesis and the clinical significance of RPS15A expression remain unclear. In this study, an evaluation of RPS15A expression in 110 surgically resected HCCs and matched tumor-adjacent normal tissues revealed an overexpression of RPS15A in HCC, which was correlated with worse survival. In addition, tumor tissue with higher RPS15A expression demonstrated a higher microvascular density (MVD). Subsequently, two HCC cell lines, Huh7 (low-level constitutive RPS15A expression) and HepG2 (high RPS15A expression) were used to further evaluate the role of RPS15A in angiogenesis. The co-culture experiment of HCC cells with endothelial cells revealed that the induced overexpression of RPS15A in Huh7 cells increased the angiogenic potential of HUVEC in a paracrine fashion; conversely, knockdown of RPS15A in HepG2 cells showed an opposite effect. Further analysis indicated that RPS15A modulated FGF signaling by enhancing Wnt/beta-catenin-mediated FGF18 expression in HCC cells. FGF18, in turn, through binding to its FGFR3 receptor on endothelial cells, can activate the AKT and ERK pathway and promotes angiogenesis in a tumor microenvironment. Our in vivo experiment further confirmed that inhibition of RPS15A expression in HCC xenografts dramatically hindered tumor growth and inhibited tumor angiogenesis. Together, our findings suggest that RPS15A promotes angiogenesis in HCCs by enhancing Wnt/beta-catenin induced FGF18 expression. The RPS15A/FGF18 pathway may be a rational target for anti-angiogenic therapy of HCC.


Subject(s)
Carcinoma, Hepatocellular/blood supply , Fibroblast Growth Factors/metabolism , Liver Neoplasms/blood supply , Neovascularization, Pathologic/pathology , Ribosomal Proteins/metabolism , Wnt1 Protein/metabolism , beta Catenin/metabolism , Adult , Aged , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Case-Control Studies , Cell Proliferation , Female , Fibroblast Growth Factors/genetics , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Lymphatic Metastasis , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Prognosis , Ribosomal Proteins/genetics , Survival Rate , Tumor Cells, Cultured , Wnt1 Protein/genetics , Xenograft Model Antitumor Assays , beta Catenin/genetics
13.
Exp Ther Med ; 14(6): 5956-5964, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29285145

ABSTRACT

Mesenchymal stem cell (MSC) therapy has emerged as a potential novel method of treating liver fibrosis. To date, bone marrow-derived MSCs (BM-MSCs) and adipose tissue-derived MSCs (AD-MSCs) have not been analyzed with respect to their ability to combat liver fibrosis. The present study aimed to compare the capabilities of BM-MSCs and AD-MSCs in the treatment of liver fibrosis. BM-MSCs and AD-MSCs were taken from male Sprague-Dawley rats and cultured. Hepatic stellate cells (HSCs) were co-cultured with either BM-MSCs or AD-MSCs, and the effects of BM-MSCs or AD-MSCs on the proliferation, activation and apoptosis of HSCs were determined. The secretion of a selected group of cytokines by BM-MSCs and AD-MSCs was measured using enzyme-linked immunosorbent assays. Using a CCl4-induced liver fibrosis animal model, the anti-inflammatory and anti-fibrotic effects of BM-MSCs or AD-MSCs against liver fibrosis in vivo were evaluated. The morphological examination and analysis of specific surface markers confirmed the successful preparation of BM-MSCs and AD-MSCs. Furthermore, the proliferation, activation and apoptosis of HSCs were significantly inhibited by BM-MSCs and AD-MSCs, with statistically greater reductions achieved by AD-MSCs compared with BM-MSCs. Direct comparison of the secretion of selected cytokines by BM-MSCs and AD-MSCs revealed that significantly higher levels of nerve growth factor and transforming growth factor-ß1 were secreted in the AD-MSC culture medium, whereas levels of vascular endothelial growth factor and interleukin-10 did not differ significantly between AD-MSCs and BM-MSCs. In vivo studies using a CCl4-induced liver fibrosis model demonstrated that inflammatory activity and fibrosis staging scores were significantly lower in the MSC-treated groups compared with controls. Although AD-MSCs improved anti-inflammatory and anti-fibrotic effects compared with BM-MSCs, these differences were not significant. Thus, the current study demonstrated that BM-MSCs and AD-MSCs are similarly effective at attenuating liver fibrosis by inhibiting the activation and proliferation of HSCs, as well as promoting the apoptosis of HSCs.

14.
Mol Med Rep ; 15(4): 1647-1653, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28260031

ABSTRACT

Hepatic stellate cell (HSC) activation serves a key role in liver fibrosis, and is associated with chronic liver diseases. Bilirubin, a product of heme degradation, has been demonstrated to have antioxidant properties. The present study investigated the effects of physiological concentrations of bilirubin on rat HSC activation. Rat HSCs were isolated and cultured for several generations to induce activation. The activated HSCs were subsequently treated with 0, 1, 10 or 20 mg/l bilirubin and assayed for parameters of cell activation. As the bilirubin concentration increased, HSCs demonstrated reduced production of reactive oxygen species, reduced protein expression levels of α­smooth muscle actin, a decreased mRNA expression ratio of tissue inhibitor of matrix metalloproteinase­1/matrix metalloproteinase­2, decreased proliferation and increased apoptosis. In conclusion, elevated bilirubin levels, within its physiological concentration range, appeared to inhibit HSC activation. These findings suggested a potential role for bilirubin in the treatment of fibrosis that requires further investigation.


Subject(s)
Bilirubin/pharmacology , Hepatic Stellate Cells/cytology , Actins , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cells, Cultured , Extracellular Matrix/drug effects , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation/drug effects , Hepatic Stellate Cells/drug effects , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism
15.
Hepatol Res ; 47(3): E187-E192, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27271941

ABSTRACT

AIM: To investigate whether ultrasound-guided percutaneous portal vein catheterization can be successfully carried out in the intrahepatic region of patients with unliquefied bacterial liver abscess (UBLA), who are subsequently treated with an injection of antibiotics. METHODS: Thirty-two UBLA patients were enrolled in this study. Among them, 13 patients were included in the experimental group; an ultrasound-guided percutaneous portal vein catheterization was undertaken in the intrahepatic region of these patients, and they also received an injection of antibiotics. The remaining 19 patients were retrospectively included in the control group; these patients only received systemic antibiotic therapy. The efficacy of intervention was compared with that of systemic treatment. RESULTS: The catheterization procedures were successful in all the patients of the experimental group. However, two cases (15.4%) developed complications postoperatively. Compared to the control group, the following parameters of the experimental group were significantly shorter/lower: (i) duration for regaining normal body temperature; (ii) time period for achieving normal white blood cell count; (iii) length of hospitalization; (iv) cases of liquefied liver abscess during follow-up; and (v) cost of hospitalization (P < 0.05). CONCLUSION: Ultrasound-guided percutaneous portal vein catheterization is a simple, minimally invasive, and effective treatment for UBLA. It must be carried out in the intrahepatic region and a subsequent injection of antibiotics must be given.

16.
Oncol Rep ; 37(1): 273-280, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27878301

ABSTRACT

Development of novel targeted therapy holds promise for conquering chemotherapy resistance, one of major hurdles in current liver cancer treatment. We found that long non-coding RNA TUC338 is involved in the development of hepatocellular carcinoma (HCC) and sorafenib resistance. HCC cell lines were transfected with siTUC338, then cell proliferation and invasion ability were investigated by MTT and Transwell assay. Sorafenib resistance HepG2 cells were generated to test the role of TUC338 in sorafenib sensitivity. Intratumoral delivering of siTUC338 was used to analyze the sorafenib treatment response in HepG2/Sor xenografts in vivo. Higher levels of TUC338 were found both in HCC tissues and cell lines, knockdown of TUC338 was accompanied with increased expression of RASAL1 in HCC cell line with increased proliferation and invasion ability, knockdown of TUC338 could activate the RASAL1 pathway and inhibit tumor growth genes by directly targeting RASAL1 3'-UTR. Furthermore, knockdown of TUC338 in HepG2 sorafenib sensitized its reaction to the treatment of sorafenib, which was accompanied by increased expression RASAL1; intratumoral delivering of siTUC338 could also restore sorafenib treatment response in HepG2/Sor xenografts in vivo. These findings provide direct evidence that the TUC338/RASAL1 axis might play an essential role in sorafenib-resistance of liver cancer cells, suggesting the signaling cohort could serve as a novel therapeutic target for the treatment of chemotherapy resistant liver cancer.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , GTPase-Activating Proteins/metabolism , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , RNA, Long Noncoding/genetics , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Nude , Niacinamide/pharmacology , Sorafenib , Xenograft Model Antitumor Assays
17.
J Exp Clin Cancer Res ; 35: 65, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-27066828

ABSTRACT

BACKGROUND: Endothelial progenitor cells (EPCs) can migrate to the tumor tissue and enhance the angiogenesis of hepatocellular carcinoma (HCC); thus, they are associated with a poor prognosis. However, the specific molecular mechanism underlying the homing of EPCs to the HCC neovasculature remains unrevealed. METHODS: Co-culture experiments of endothelial progenitor cells with HCC cells with modulation of EphA1 were performed in vitro. Using EPCs as angiogenic promoters by injecting them into HCC xenograft-bearing nude mice via their tail veins to test homing ability of EPCs changed according to different EphA1 level in HCC xenograft. RESULTS: In this study, we found that the up-regulation of EphA1 expression in HCC cells could affect not only the chemotaxis of EPCs to tumor cells and endothelial cells (ECs) but also the tube formation ability of EPCs in a paracrine fashion. Further, we revealed that the increased expression of EphA1 in HCC cells led to an increased SDF-1 concentration in the tumor microenvironment, which in turn activated the SDF-1/CXCR4 axis and enhanced the recruitment of EPCs to HCC. In addition, the EphA1-activated SDF-1 expression and secretion was partially mediated by the PI3K and mTOR pathways. In vivo experiments demonstrated that blocking EphA1/SDF-1/CXCR4 signaling significantly inhibited the growth of HCC xenografts. Using immunohistochemistry and immunofluorescence assays, we verified that the inhibition of tumor angiogenesis was at least partially caused by the decreased number of EPCs homing to tumor tissue. CONCLUSIONS: Our findings indicate that targeting the EphA1/SDF-1 signaling pathway might be a therapeutic anti-angiogenesis approach for treating HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Chemokine CXCL12/metabolism , Endothelial Cells/cytology , Liver Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Receptor, EphA1/metabolism , Receptors, CXCR4/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement , Chemokine CXCL12/genetics , Coculture Techniques , Endothelial Cells/transplantation , Human Umbilical Vein Endothelial Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Receptors, CXCR4/genetics , Signal Transduction , Tumor Microenvironment
18.
Hepatobiliary Pancreat Dis Int ; 14(2): 215-21, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25865696

ABSTRACT

BACKGROUND: Pancreatic stellate cells (PSCs) play a critical role in the development of pancreatic fibrosis. In this study we used a novel method to isolate and culture rat PSCs and then investigated the inhibitory effects of adipose-derived stem cells (ADSCs) on activation and proliferation of PSCs. METHODS: Pancreatic tissue was obtained from Sprague-Dawley rats for PSCs isolation. Transwell cell cultures were adopted for co-culture of ADSCs and PSCs. PSCs proliferation and apoptosis were determined using CCK-8 and flow cytometry, respectively. alpha-SMA expressions were analyzed using Western blotting. The levels of cytokines [nerve growth factor (NGF), interleukin-10 (IL-10) and transforming growth factor-beta1 (TGF-beta1)] in conditioned medium were detected by ELISA. Gene expression (MMP-2, MMP-9 and TIMP-1) was analyzed using qRT-PCR. RESULTS: This method produced 17.6+/-6.5X10(3) cells per gram of the body weight with a purity of 90%-95% and a viability of 92%-97%. Co-culture of PSCs with ADSCs significantly inhibited PSCs proliferation and induced PSCs apoptosis. Moreover, alpha-SMA expression was significantly reduced in PSCs+ADSCs compared with that in PSC-only cultures, while expression of fibrinolytic proteins (e.g., MMP-2 and MMP-9) was up-regulated and anti-fibrinolytic protein (TIMP-1) was down-regulated. In addition, NGF expression was up-regulated, but IL-10 and TGF-beta1 expressions were down-regulated in the co-culture conditioned medium compared with those in the PSC-only culture medium. CONCLUSIONS: This study provided an easy and reliable technique to isolate PSCs. The data demonstrated the inhibitory effects of ADSCs on the activation and proliferation of PSCs in vitro.


Subject(s)
Adipose Tissue/cytology , Apoptosis , Cell Proliferation , Pancreatic Stellate Cells/physiology , Stem Cells , Actins/metabolism , Animals , Coculture Techniques , Culture Media, Conditioned , Down-Regulation/genetics , Gene Expression , Interleukin-10/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Nerve Growth Factor/metabolism , Rats , Rats, Sprague-Dawley , Tissue Inhibitor of Metalloproteinase-1/genetics , Transforming Growth Factor beta1/metabolism , Up-Regulation/genetics
19.
World J Surg Oncol ; 13: 52, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25890096

ABSTRACT

BACKGROUND: Bone marrow-derived stem cells (BMSCs) are locally adjacent to the tumor tissues and may interact with tumor cells directly. The purpose of this study was to explore the effects of BMSCs on the proliferation and invasion of osteosarcoma cells in vitro and the possible mechanism involved. METHODS: BMSCs were co-cultured with osteosarcoma cells, and CCK-8 assay was used to measure cell proliferation. The ELISA method was used to determine the concentration of stromal cell-derived factor-1 (SDF-1) in the supernatants. Reverse transcription polymerase chain reaction (RT-PCR) was performed to detect the expression of CXCR4 in osteosarcoma cells and BMSCs. Matrigel invasion assay was performed to measure tumor cell invasion. RESULTS: SDF-1 was detected in the supernatants of BMSCs, but not in osteosarcoma cells. Higher CXCR4 mRNA levels were detected in the osteosarcoma cell lines compared to BMSCs. In addition, conditioned medium from BMSCs can promote the proliferation and invasion of osteosarcoma cells, and AMD3100, an antagonist for CXCR4, can significantly downregulate these growth-promoting effects. CONCLUSIONS: BMSCs can promote the proliferation and invasion of osteosarcoma cells, which may involve the SDF-1/CXCR4 axis.


Subject(s)
Bone Marrow Cells/pathology , Bone Neoplasms/pathology , Cell Movement , Cell Proliferation , Mesenchymal Stem Cells/pathology , Osteosarcoma/pathology , Apoptosis , Blotting, Western , Bone Marrow Cells/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Flow Cytometry , Humans , Immunoenzyme Techniques , Mesenchymal Stem Cells/metabolism , Osteosarcoma/genetics , Osteosarcoma/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
20.
J Formos Med Assoc ; 114(2): 130-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25678175

ABSTRACT

BACKGROUND/PURPOSE: Previous studies suggested that mesenchymal stem cells may ameliorate fibrogenesis through the inhibition of hepatic stellate cells (HSCs) activation. This study aimed to investigate whether adipose derived mesenchymal stem cells (ADSCs) could modulate the activation of HSCs and contribute to the recovery of liver fibrogenesis. METHODS: ADSCs and HSCs were isolated from Sprague-Dawley rats and co-cultured using a transwells insert. Cell proliferation, apoptosis and smooth muscle α-actin (α-SMA) expression in HSCs were examined. Rats were injected with CCl4 to induce liver fibrogenesis. After injection of ADSCs through portal vein, the rats were examined for pathological changes in the liver. α-SMA expression and hydroxyproline content in the liver and serum levels of collagen III and hyaluronic acid was detected. RESULTS: After co-culturing for 72 h, the proliferation and activation of HSCs was inhibited by ADSCs and the apoptosis of HSCs was promoted by ADSCs. Transplantation of ADSCs inhibited liver fibrogenesis in the rats. CONCLUSION: ADSCs inhibit the proliferation and activation of HSCs in vitro and inhibit liver fibrogenesis in rat model, suggesting the potential application of ADSCs in liver fibrogenesis therapy.


Subject(s)
Hepatic Stellate Cells/cytology , Liver Cirrhosis/therapy , Liver/pathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Actins/metabolism , Adipose Tissue/cytology , Animals , Apoptosis , Carbon Tetrachloride , Cell Proliferation , Cells, Cultured , Liver Cirrhosis/chemically induced , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL