Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
BMC Surg ; 22(1): 21, 2022 Jan 22.
Article in English | MEDLINE | ID: mdl-35065645

ABSTRACT

BACKGROUND: The choice of surgical strategy for patients with rectal gastrointestinal stromal tumor (GIST) remains controversial. This study aims to address whether the surgical procedure [local excision (LE) vs. radical excision (RE)] influences the survival outcomes. METHODS: The information of the patients recruited in this study was obtained from the Surveillance, Epidemiology, and End Results (SEER) database. A survival curve was used to evaluate the differences in cancer-specific survival (CSS). RESULTS: No significant difference was detected in the CSS between the LE and RE groups. Also, no significant differences were observed in the CSS between the two groups with respect to different T classification, N classification, tumor differentiation, tumor size, regional LN surgery, age, gender, race, chemotherapy, and radiotherapy. The T classification and age were independent prognostic factors in rectal GIST patients. CONCLUSIONS: LE and RE have similar survival time after surgery, and LE could be considered as an effective surgical approach for rectal GIST.


Subject(s)
Digestive System Surgical Procedures , Gastrointestinal Stromal Tumors , Rectal Neoplasms , Databases, Factual , Gastrointestinal Stromal Tumors/surgery , Humans , Neoplasm Staging , Rectal Neoplasms/pathology , Rectal Neoplasms/surgery , Rectum/pathology
3.
BMC Cancer ; 21(1): 1138, 2021 Oct 23.
Article in English | MEDLINE | ID: mdl-34688251

ABSTRACT

BACKGROUND: Compared to conventional adenocarcinoma (CA), mucin-producing adenocarcinoma (MPA) is an uncommon histological subtype and is usually separated from other histological types and has been evaluated separately. The objective was to compare the clinicopathological characteristics and survivals of MPA with CA. METHODS: We retrospectively analyzed 1515 MPA patients in SEER database. Log-rank tests and KM survival curves were applied to determine the differences in overall survival (OS) and cancer specific survival (CSS) time. RESULTS: No significant differences were noted in OS and CSS time. The MPA patients who were treated with surgery and chemotherapy exhibited longer OS and CSS time periods than those without treatment. MPA patients treated with radiotherapy exhibited similar OS and CSS time with those without radiotherapy. MPA was not a prognostic factor of survival. CONCLUSIONS: MPA was a rare histological type of gastric cancer. Patients with MPA exhibited similar prognosis with those with CA. Surgery and chemotherapy were effective treatments for patients with MPA.


Subject(s)
Adenocarcinoma/mortality , Adenocarcinoma/physiopathology , Gastric Mucins/metabolism , SEER Program/standards , Stomach Neoplasms/mortality , Stomach Neoplasms/physiopathology , Female , Humans , Male , Middle Aged , Survival Analysis , Treatment Outcome
4.
Cancer Manag Res ; 13: 4777-4790, 2021.
Article in English | MEDLINE | ID: mdl-34168497

ABSTRACT

BACKGROUND: Mucinous adenocarcinoma (MA) is a subtype of colorectal cancer (CRC) associated with a higher incidence of local extension and worse survival compared to non-mucinous adenocarcinoma, but few studies have investigated surgery-related predictors for recurrence of MA. Therefore, we aimed to elucidate the predictors for local recurrence and remote metastasis of MA after surgery. PATIENTS AND METHODS: This study retrospectively analyzed 162 patients with mucinous colorectal adenocarcinoma (MAC) after radical resection. Analysis variables included demographics, clinical indicators, pathologic stage, surgical procedure, adjuvant therapy, and recurrence. Univariate and multivariate analyses were performed to investigate the risk factors for local and distant tumor relapse. RESULTS: A total of 162 patients (86 male) with a mean age of 58.26 years were included; 70.37% of patients had colonic tumors, and 29.63% had rectal tumors. The 5-year disease-free survival (DFS) rates for these patients were as follows: 100% for TNM stage I, 71.2% for stage II, and 47.8% for stage III. Five-year DFS rates of MAC, colonic and rectal MA were 62.0%, 65.8%, and 51.7%, respectively. Local recurrence occurred in 38 patients and distant metastasis in 33 patients. In univariate analysis, predictors for local recurrence of MAC were intraoperative blood loss, intraoperative transfusion, and N2 stage; and predictors for distant metastasis were male sex, CA199, CEA, intraoperative blood loss, T4 stage, and N2 stage. In multivariate analysis, predictors for local recurrence of MAC were intraoperative transfusion (P=0.04, OR=4.175) and N2 stage (P=0.000, OR=5.291), and predictors for distant metastasis were male sex (P=0.049, OR=2.410), CA199 (P=0.02, OR=1.003), and T4 stage (P=0.007, OR=4.006). CONCLUSION: Intraoperative transfusion and N2 stage were significant predictors for local recurrence. Male sex, CA199, and T4 stage were significant predictors for distant metastasis. Knowledge of the risk factors for postoperative recurrence provides a basis for logical approaches to treatment and follow-up of MAC.

5.
J Laparoendosc Adv Surg Tech A ; 31(6): 638-647, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33151807

ABSTRACT

Background: Mucinous colorectal adenocarcinoma (MAC) has a higher incidence of local extension, leading to lower overall resection rates. Few studies have investigated the outcomes of laparoscopic surgery for MACs to date. Therefore, we aimed to elucidate the validity of laparoscopic surgery for mucinous adenocarcinoma (MAC). Methods: This study analyzed short-term and long-term outcomes between laparoscopic and open surgery for MACs from 2008 to 2018. Multivariate analyses were used to define prognostic factors of overall survival (OS) and disease-free survival (DFS). Results: Patients in the laparoscopy (LAP) group had significantly less blood loss, fewer days to first flatus and to diet, and shorter length of hospital stay. The 3-year and 5-year DFS rates for all stages combined were 65.7% and 62.5% in the LAP group compared with 60.5% and 57.6% in the open (OPEN) surgery group (P = .521). The 3-year and 5-year OS rates for all stages combined were 72.3% and 67.3% in the LAP group compared with 72.6% and 67.8% in the OPEN group (P = .934). OS and DFS in stage II, stage III, and pathological T4 (pT4) stage patients who underwent laparoscopic surgery did not differ from patients who underwent open surgery. Multivariate analysis showed that stage pT4, pN2, and carcinoembryonic antigen (CEA) were significant predictors of OS. Independent factors, including intraoperative blood transfusion, stage pT4, pN2, CEA, and CA19-9, carbohydrate antigen 19-9, have a great effect on DFS. Conclusions: Laparoscopic surgery is a safe and feasible option for mucinous colorectal AC, which provides faster postoperative recovery and less intraoperative blood loss.


Subject(s)
Adenocarcinoma, Mucinous/surgery , Colorectal Neoplasms/surgery , Laparoscopy , Adenocarcinoma, Mucinous/blood , Adenocarcinoma, Mucinous/secondary , Adult , Aged , Antigens, Tumor-Associated, Carbohydrate/blood , Blood Loss, Surgical , Blood Transfusion , Carcinoembryonic Antigen/blood , Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Disease-Free Survival , Female , Gastrointestinal Tract/physiopathology , Humans , Length of Stay , Male , Middle Aged , Neoplasm Staging , Recovery of Function , Survival Rate , Treatment Outcome
6.
Future Oncol ; 17(10): 1185-1195, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33289395

ABSTRACT

Aims: To addresses whether surgical procedure (proximal gastrectomy [PG] vs total gastrectomy [TG]) influences survival outcomes. Methods: Patients were selected from Surveillance, Epidemiology and End Results Program (SEER) database. Survival curve was used to evaluate the differences in overall survival (OS) and cancer-specific survival (CSS). Results: No significant difference was detected in OS and CSS time between PG and TG groups. Also, no significant differences were observed in OS and CSS times between the two groups with respect to clinical stage, tumor stage, node stage, age, gender and tumor differentiation. Tumor differentiation, tumor size, tumor stage, node stage and age were independent prognostic factors in patients with proximal gastric cancer. Conclusions: TG was not necessary for proximal gastric cancer patients, and PG may be considered as an ideal surgery approach.


Subject(s)
Gastrectomy , Stomach Neoplasms/epidemiology , Stomach Neoplasms/surgery , Disease Management , Female , Gastrectomy/methods , Health Care Surveys , Humans , Male , Prognosis , SEER Program , Stomach Neoplasms/pathology , Treatment Outcome
7.
Oncol Lett ; 17(3): 3314-3322, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30867765

ABSTRACT

The current study aimed to develop multiple diagnosis models for colorectal cancer (CRC) based on data from The Cancer Genome Atlas database and analysis with artificial neural networks in order to enhance CRC diagnosis methods. A genetic algorithm and mean impact value were used to select genes to be used as numerical encoded parameters to reflect cancer metastasis or aggression. Back propagation and learning vector quantization neural networks were used to build four diagnosis models: Cancer/Normal, M0/M1, carcinoembryonic antigen (CEA) <5/≥5 and Clinical stage I-II/III-IV. The performance of each model was evaluated by predictive accuracy (ACC), the area under the receiver operating characteristic curve (AUC) and a 10-fold cross-validation test. The ACC and AUC of the Cancer/Normal, M0/M1, CEA and Clinical stage models were 100%, 1.000; 87.14%, 0.670; 100%, 1.000; and 100%, 1.000, respectively. The 10-fold cross-validation test of the ACC values and sensitivity for each test were 93.75-99.39%, 1.0000; 80.58-88.24%, 0.9286-1.0000; 67.21-92.31%, 0.7091-1.0000; and 59.13-68.85%, 0.6017-0.6585, respectively. The diagnosis models developed in the current study combined gene expression profiling data and artificial intelligence algorithms to create tools for improved diagnosis of CRC.

8.
Cancer Lett ; 450: 1-13, 2019 05 28.
Article in English | MEDLINE | ID: mdl-30776481

ABSTRACT

PD-L1 is critical for tumor cell escape from immune surveillance by inhibiting T cell function via the PD-1 receptor. Accumulating evidence demonstrates that anti-PD-L1 monoclonal antibodies might potently enhance antitumor effects in various tumors, but the effect of PD-L1 on colorectal cancer stem cells (CSCs) remains unclear. We observed high PD-L1 expression in CD133+CD44+ colorectal CSCs and CSC-enriched tumorspheres. Altering PD-L1 expression promoted colorectal CSC self-renewal by increasing the expression of stemness genes, the CD133+CD44+ cell population sizes and the ability to form tumorspheres. Additionally, PD-L1 expression was markedly increased in chemoresistant colorectal cancer (CRC) cells in vitro and in vivo. More importantly, PD-L1 enhanced CRC cell tumorigenicity in nude mice; the inoculation of 1 × 104 cells resulted in high tumor formation efficiency. Mechanistically, PD-L1 directly interacted with HMGA1, and HMGA1 upregulation by PD-L1 activated HMGA1-dependent pathways, including the PI3K/Akt and MEK/ERK pathways, and promoted CSC expansion. HMGA1 downregulation rescued the PD-L1-induced phenotypes, highlighting the role of HMGA1 in PD-L1-mediated colorectal CSC self-renewal. Moreover, PD-L1 expression was correlated with the expression of CSC markers and HMGA1 in clinical CRC specimens. Thus, PD-L1 could crucially contribute to the maintenance of CSC self-renewal by activating HMGA1-dependent signaling pathways.


Subject(s)
B7-H1 Antigen/metabolism , Colorectal Neoplasms/metabolism , HMGA1a Protein/metabolism , Neoplastic Stem Cells/metabolism , Animals , B7-H1 Antigen/biosynthesis , Cisplatin/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , HCT116 Cells , HT29 Cells , Humans , Immunohistochemistry , Mice , Mice, Nude , Neoplastic Stem Cells/pathology , Signal Transduction , Xenograft Model Antitumor Assays
9.
Oncogene ; 38(2): 164-179, 2019 01.
Article in English | MEDLINE | ID: mdl-30076414

ABSTRACT

Long noncoding RNAs (lncRNAs) are implicated in human cancer, but their mechanisms of action are largely unknown. In this study, we investigated lncRNA alterations that contribute to colorectal cancer (CRC) through microarray expression profiling in CRC patient samples. Here, we report that the CRC-associated lncRNA PVT1-214 is a key regulator of CRC development and progression; patients with high PVT1-214 expression had a shorter survival and poorer prognosis. In vitro and in vivo investigation of the role of PVT1-214 revealed a complex integrated phenotype affecting cell growth, stem-like properties, migration, and invasion. Furthermore, using RNA pull-down and mass spectrometry, we found that Lin28 (also known as Lin28A), a highly conserved RNA-binding protein, is associated with PVT1-214. Strikingly, we found that PVT1-214 not only upregulated Lin28 protein expression in CRC cells by stabilizing Lin28, but also participated in crosstalk with Lin28 mRNA through competition for miR-128 binding, imposing an additional level of post-transcriptional regulation. In addition, we further show that PVT1-214 repressed expression of let-7 family miRNAs, which was abrogated by Lin28 knockdown. Taken together, our findings support a model in which the PVT1-214/Lin28/let-7 axis serves as a critical regulator of CRC pathogenesis, which may simulate a new direction for CRC therapeutic development.


Subject(s)
Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA-Binding Proteins/genetics , Animals , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology
10.
Mol Cancer ; 17(1): 121, 2018 08 13.
Article in English | MEDLINE | ID: mdl-30103756

ABSTRACT

Genome-scale CRISPR-Cas9 Knockout Screening was applied to investigate novel targets in imatinib-resistant gastrointestinal stromal tumor (GIST). 20 genes and 2 miRNAs have been selected by total reads of sgRNA and sgRNA diversity, which has been further validated in imatinib-resistant GIST cells by CCK8 and qPCR analysis. Our study has finally revealed 9 genes (DBP, NR3C1, TCF12, TP53, ZNF12, SOCS6, ZFP36, ACYP1, and DRD1) involved in imatinib-resistant GIST-T1 cells. TP53 and SOCS6 may be the most promising candidate genes for imatinib-resistance due to the possible signaling pathway, such as apoptosis pathway and Wnt signaling pathway, JAK-STAT signaling pathway. It is necessary to perform more studies to discover novel targets in imatinib-resistant GIST, including DBP, NR3C1, TCF12, ZNF12, ZFP36, ACYP1 and DRD1.


Subject(s)
Drug Resistance, Neoplasm , Gastrointestinal Neoplasms/genetics , Gastrointestinal Stromal Tumors/genetics , Gene Regulatory Networks , CRISPR-Cas Systems , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Imatinib Mesylate , Signal Transduction
11.
Int J Oncol ; 53(3): 1227-1236, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29956750

ABSTRACT

Colorectal cancer (CRC) is the third most common type of cancer; however, the molecular mechanisms underlying colorectal tumor metastasis and growth remain elusive. Recently, accumulating evidence has indicated that long non­coding RNAs (lncRNAs) play a critical role in CRC progression and metastasis; however, the biological role and clinical significance of lncRNA 00152 (lnc00152) in CRC remains largely unknown. Thus, in this study, lnc00152 expression was measured in 80 human CRC tissue samples, 40 non­cancerous tissue samples, and 3 CRC cell lines (SW480, SW620 and LoVo) using RT­qPCR. We examined the effects of lnc00152 on CRC cells following transfection with lnc00152 overexpression plasmid or respective siRNA in vitro and in vivo. Luciferase assays revealed the mechanism driving competitive endogenous RNA (ceRNA). We identified that lnc00152 was aberrantly overexpressed in colorectal tumors and cancer cells and that lnc00152 was modulated by miRNA­206. lnc00152 overexpression enhanced the proliferative and invasive ability of CRC cells in vitro, promoted tumor growth in vivo, and was associated with the shorter overall survival of patients with CRC. In addition, lnc00152 overexpression promoted epithelial-mesenchymal transition (EMT) and increased neuropilin­1 (NRP1) expression in the CRC cells. By contrast, lnc00152 silencing exerted a counteractive effect. Collectively, these findings demonstrate the critical role of lnc00152 in tumor growth and progression in CRC, and identify a novel therapeutic target associated with CRC development and progression.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Neuropilin-1/genetics , RNA, Long Noncoding/metabolism , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Kaplan-Meier Estimate , Male , Mice , Mice, Nude , MicroRNAs/genetics , Middle Aged , Mutagenesis, Site-Directed , Neoplasm Invasiveness/genetics , Neuropilin-1/metabolism , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , Xenograft Model Antitumor Assays
12.
Cell Physiol Biochem ; 46(2): 829-846, 2018.
Article in English | MEDLINE | ID: mdl-29627822

ABSTRACT

BACKGROUND/AIMS: Gambogic acid (GA), the main active compound of Gamboge hanburyi, has been reported to be a potential novel antitumor drug. Whether GA inhibits putative cancer stem cells (CSCs), which are considered to be the major cause of cancer treatment failure, remains largely unknown. This study investigated whether GA inhibits the CSCs of colorectal cancer (CRC) and its possible mechanisms. METHODS: We performed CCK8 and tumor sphere formation assays, percentage analysis of both side population and CD133+CD44+ cells, and the detection of stem cells markers, in order to assess the role of GA in inhibiting the stem celllike features of CRC. An mRNA microarray was performed to identify the downstream gene affected by GA and rescue assays were performed to further clarify whether the downstream gene is involved in the GA induced decrease of the stem cell-like CRC population. CRC cells were engineered with a CSC detector vector encoding GFP and luciferase (Luc) under the control of the Nanog promoter, which were utilized to investigate the effect of GA on putative CSC in human tumor xenograft-bearing mice using in vivo bioluminescence imaging. RESULTS: Our results showed that GA significantly reduced tumor sphere formation and the percentages of side population and CD133+CD44+ cells, while also decreasing the expression of stemness and EMT-associated markers in CRC cells in vitro. GA killed stem-like CRC cells by upregulating the expression of ZFP36, which is dependent on the inactivation of the EGFR/ ERK signaling pathway. GFP+ cells harboring the PNanog-GFP-T2A-Luc transgene exhibited CSC characteristics. The in vivo results showed that GA significantly inhibited tumor growth in nude mice, accompanied by a remarkable reduction in the putative CSC number, based on whole-body bioluminescence imaging. CONCLUSION: These findings suggest that GA significantly inhibits putative CSCs of CRC both in vitro and in vivo by inhibiting the activation of the EGFR/ ERK/ZFP36 signaling pathway and may be an effective drug candidate for anticancer therapies.


Subject(s)
Apoptosis/drug effects , Tristetraprolin/metabolism , Up-Regulation/drug effects , Xanthones/toxicity , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibronectins/genetics , Fibronectins/metabolism , HCT116 Cells , Humans , Mice , Mice, Nude , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction/drug effects , Transplantation, Heterologous , Tristetraprolin/genetics , Xanthones/therapeutic use
13.
Oncol Lett ; 15(3): 2953-2961, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29435024

ABSTRACT

Enhancer of zeste homolog 2 (EZH2), the critical component of polycomb group protein family, has been demonstrated to be overexpressed in various types of human cancer, including hepatocellular carcinoma, breast, bladder and lung cancer. The mechanism of how EZH2 promotes oncogenesis has also been well studied. However, little is known about the role of EZH2 in colorectal cancer (CRC). The main purpose of the present study was to analyze the association between EZH2 expression and the clinicopathological features of CRC. Therefore, the mRNA and protein expression levels were analyzed in tumor tissues and adjacent non-cancerous tissues by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis. The expression of EZH2 was demonstrated to be significantly increased in tumor tissues compared with adjacent noncancerous tissues, according to the results of western blot analysis and RT-qPCR in the majority of cases. Patients with low EZH2 expression had a longer overall survival rate compared with those with high EZH2 expression. An analysis of the association between clinicopathological features and EZH2 expression indicated that high EZH2 expression was significantly associated with tumor stage, tumor size, histological differentiation and lymph node metastasis. Multivariate analysis demonstrated that high EZH2 expression was an independent predictor of overall survival. In conclusion, to the best of our knowledge, the data presented in the present study is the first to indicate that EZH2 is upregulated in CRC and may serve as a predictor of poor outcome for patients with CRC.

14.
Exp Ther Med ; 14(4): 3057-3061, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28912857

ABSTRACT

Transcription factor activating enhancer binding protein 4 (TFAP4) is an important regulator in the genesis and progression of human cancers. Overexpression of TFAP4 has been found to be correlated with several malignancies. The present study assessed the clinical importance of TFAP4 in colorectal cancer (CRC). First, immunohistochemistry was used to analyze TFAP4 expression and the association of TFAP4 expression with clinicopathological features on a tissue microarray containing 208 CRC patients. The results revealed that TFAP4 protein expression was significantly upregulated in CRC tissues compared with that in normal colon tissues (P<0.001). Of note, statistical analysis revealed that TFAP4 expression was significantly correlated with a high pathological grade (P=0.034), advanced clinical stage (P=0.024), enhanced tumor invasion (P=0.002) and lymph node metastasis (P=0.041). In addition, the Cancer Genome Atlas dataset further validated that TFAP4 mRNA levels were increased in CRC with advanced clinical stage (P=0.026), lymph node metastasis (P=0.018) and vascular invasion (P=0.046). Kaplan-Meier survival analysis demonstrated that CRC patients with high TFAP4 expression had shorter overall survival compared with those with low TFAP4 expression (P=0.011). Importantly, overexpression of TFAP4 was a valuable independent prognostic factor for CRC patients (hazard ratio, 8.200; 95% confidence interval, 1.838-36.591; P=0.006). In summary, TFAP4 may have an important role in CRC progression and upregulation of TFAP4 may be a predictor of poor prognosis for CRC patients.

15.
Exp Ther Med ; 13(2): 662-668, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28352348

ABSTRACT

Chemotherapy using 5-fluorouracil (5-FU) for colorectal cancer (CRC) has low specificity and response rates, leading to severe side effects. Gambogic acid (GA), a traditional Chinese medicine, has multi-targeted anticancer effects, including growth inhibition and apoptosis induction. However, it is unclear whether a combination of 5-FU and GA has synergistic anticancer effects in CRC cells. In this study, SW480 and HCT116 human CRC cells and human intestinal epithelial cells (IECs) were treated with different concentrations of 5-FU, GA or 5-FU+GA. A Cell Counting kit-8 assay was conducted to quantify cell proliferation. The combination index (CI) was calculated and the median-effect principle was applied to analyze the interaction between 5-FU and GA. Flow cytometry was used to determine the percentage of cells undergoing apoptosis. Reverse transcription-quantitative polymerase chain reaction and western blotting were applied to measure P53, survivin and thymidylate synthase (TS) mRNA and protein levels. It was found that 5-FU+GA more pronouncedly inhibited cell growth and induced apoptosis, compared with either monotherapy. CI values <1 indicated the synergistic effects of the drugs. 5-FU+GA further decreased P53, survivin and TS mRNA and protein levels in the two CRC cell lines compared with single drugs, whereas increased P53 protein levels were observed in HCT116 cells. Moreover, 5-FU+GA did not increase cytotoxicity to IECs. These results demonstrate that GA enhances the anticancer effects of 5-FU on CRC cells. Combined treatment with 5-FU and GA is effective and safe for CRC cells, and may become a promising chemotherapy treatment.

16.
Oncotarget ; 7(49): 81156-81171, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27835604

ABSTRACT

High levels of angiogenesis, metastasis and chemoresistance are major clinical features of colorectal cancer (CRC), a lethal disease with a high incidence worldwide. Aberrant activation of Wnt/ß-catenin pathway contributes to CRC progression. However, little is known about regulatory mechanisms of the ß-catenin activity in cancer progression. Here we report that Gankyrin was markedly upregulated in primary tumor tissues from CRC patients and was associated with poor survival. Moreover, we demonstrated that overexpressing Gankyrin promoted, while knockdown of Gankyrin impaired, the aggressive phenotype of proliferation, angiogenesis, chemoresistance and metastasis of CRC cells both in vitro and in vivo. Importantly, we found a unique molecular mechanism of Gankyrin in CRC cells signaling transduction, that regulated the cross-talk between PI3K/Akt and Wnt/ß-catenin signaling pathways, sustaining PI3K/GSK-3ß/ß-catenin signal activation in CRC. Therefore, these findings not only reveal a mechanism that promotes aggressiveness and progression in CRC, but also provide insight into novel molecular targets for antitumor therapy in CRCs.


Subject(s)
Colorectal Neoplasms/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins/metabolism , beta Catenin/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Cell Movement , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Computational Biology , Databases, Genetic , Drug Resistance, Neoplasm , Female , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Male , Mice, Inbred BALB C , Middle Aged , Neoplasm Invasiveness , Neovascularization, Pathologic , Phenotype , Proteasome Endopeptidase Complex/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Time Factors , Transfection , Wnt Signaling Pathway
17.
Zhonghua Wei Chang Wai Ke Za Zhi ; 19(6): 695-701, 2016 Jun.
Article in Chinese | MEDLINE | ID: mdl-27353107

ABSTRACT

OBJECTIVE: To investigate the effect and its possible mechanism of interleukin-17 (IL-17) on invasion and metastasis of human colon cancer cells. METHODS: IL-17 was added into the culture media of human colon cancer cells SW480 and LOVO. Cells were divided into 4 groups: SW480 control group (SW480 cells), LOVO control group (LOVO cells), SW480 experiment group (50 µg/L IL-17+SW480 cells), and LOVO experiment group (50 µg/L IL-17+LOVO cells). Cell growth was measured by CCK-8 assay. The proliferation rate(%)=[(Aexperiment group-Ablank)/(Acontrol group-Ablank)]×100%). The ability of cell invasion and migration was measured by transwell assay. Real time-PCR was used to detect mRNA expression of VEGF and MMP-9. Western blot was performed to detect protein expression of STAT3, p-STAT3, VEGF and MMP-9. Enzyme-linked immunosorbent assay (ELISA) was applied to measure the protein content of VEGF and MMP-9 in the supernatant. RESULTS: After cultivation for 24, 48 and 72 hours, CCK-8 assay revealed that the proliferation rate of SW480 was 1.18%±0.07%, 1.42%±0.09%, and 1.62%±0.08%; the proliferation rate of LOVO was 1.13%±0.02%, 1.32%±0.05% and 1.73%±0.02% in experiment group. Transwell experiments showed that after cultivation with IL-17 for 24 hours, number of invasion cell in experimental groups (SW480: 34.00±0.45, LOVO: 41.60±0.51) was higher as compared to corresponding control groups (SW480: 4.53±0.14; LOVO: 3.67±0.33) with significant differences (SW480: t=-76.026, P=0.001; LOVO: t=-81.580, P=0.005). The number of migration cell in experimental groups (SW480: 36.40±0.51, LOVO: 46.40±0.68) was higher as compared to corresponding control groups (SW480: 7.83±0.69; LOVO: 6.67±0.48) with significant differences (SW480: t=-51.542, P=0.003; LOVO: t=-49.265, P=0.005). Real-time PCR results revealed that after cultivation with IL-17 for 24 hours, VEGF and MMP-9 mRNA relative expression levels in experimental groups (SW480: VEGF:1.53±0.12, MMP-9: 2.44±0.23; LOVO: VEGF: 2.96±0.35, MMP-9: 3.38±0.55) were higher than those in control groups (both 1) with significant differences (VEGF: t=3.799, P=0.043; MMP-9: t=5.254, P=0.039). Western blot illustrated that after cultivation with IL-17 for 24 hours, STAT3, p-STAT3, VEGF and MMP-9 proteins relative expression levels in experimental groups were significantly higher that those in control groups (SW480:STAT3: t=3.233, P=0.023; p-STAT3: t=3.954, P=0.032; VEGF: t=3.201, P=0.025; MMP-9: t=3.154, P=0.029; LOVO: STAT3: t=3.788, P=0.012; p-STAT3: t=2.662, P=0.040; VEGF: t=4.118, P=0.035; MMP-9: t=4.268, P=0.030). ELISA indicated that content of VEGF and MMP-9 in the supernatant of experimental groups (SW480: VEGF 5 491.41±63.22, MMP-9: 21.43±1.35. LOVO: VEGF: 8 631.46±129.59, MMP-9: 178.32±3.20) were higher than those in control groups (SW480: VEGF:4 456.32±87.56, MMP-9:18.57±2.44. LOVO: VEGF: 8 122.38±108.66, MMP-9: 163.22±6.89) with significant differences (SW480: VEGF: t=6.993, P=0.037; MMP-9: t=5.587, P=0.040. LOVO: VEGF: t=7.013, P=0.044; MMP-9: t=6.762, P=0.043). CONCLUSION: IL-17 may be able to activate STAT3 signal transduction pathway in vitro through up-regulation of VEGF and MMP-9 expression, thereby enhancing the invasion and migration of colon cancer SW480 and LOVO cells.


Subject(s)
Colonic Neoplasms/pathology , Interleukin-17/pharmacology , Signal Transduction , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Real-Time Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...