Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
1.
Cancer Biomark ; 30(4): 395-406, 2021.
Article in English | MEDLINE | ID: mdl-33386794

ABSTRACT

BACKGROUND: Glycolysis was an essential driver of chemo-resistance in colorectal cancer (CRC), albeit with limited molecular explanations. OBJECTIVE: We strived to elucidate the involvement of lncRNA XIST/miR-137/PKM axis in chemo-tolerance and glycolysis of CRC. METHODS: Altogether 212 pairs of tumor tissues and adjacent normal tissues were collected from CRC patients. Moreover, human CRC epithelial cell lines, including HT29, SW480, SW620 and LoVo, were purchased in advance, and their activity was estimated after transfection of si-XIST or miR-137 mimic. Furthermore, 5-FU/cisplatin-resistance of CRC cells was determined through MTT assay, and glycolytic potential of CRC cells was appraised based on oxygen consumption rate (OCR) and extracellular acidification rate (ECAR). RESULTS: Highly-expressed XIST were predictive of severe symptoms and unfavorable 3-year survival of CRC patients (P< 0.05). Besides, silencing of XIST not only diminished proliferative, migratory and invasive power of CRC cells (P< 0.05), but also enhanced sensitivity of CRC cells responding to 5-FU/cisplatin (P< 0.05). Glycolytic potency of CRC cells was also undermined by si-XIST, with decreased maximal respiration and maximal glycolytic capacity in the si-XIST group as relative to NC group (P< 0.05). Nevertheless, miR-137 mimic attenuated the facilitating effect of pcDNA3.1-XIST on proliferation, migration, invasion, 5-FU/cisplatin-resistance and glycolysis of CRC cells (P< 0.05). Ultimately, ratio of PKM2 mRNA and PKM1 mRNA, despite being up-regulated by pcDNA3.1-XIST, was markedly lowered when miR-137 mimic was co-transfected (P< 0.05). CONCLUSIONS: LncRNA XIST/miR-137 axis reinforced glycolysis and chemo-tolerance of CRC by elevating PKM2/PKM1 ratio, providing an alternative to boost chemo-therapeutic efficacy of CRC patients.


Subject(s)
Carrier Proteins/metabolism , Colorectal Neoplasms/metabolism , Membrane Proteins/metabolism , MicroRNAs/metabolism , Pyruvate Kinase/metabolism , RNA, Long Noncoding/metabolism , Thyroid Hormones/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Female , Glycolysis , HT29 Cells , Humans , MicroRNAs/genetics , Pyruvate Kinase/genetics , RNA, Long Noncoding/genetics , Transfection , Thyroid Hormone-Binding Proteins
2.
Front Med (Lausanne) ; 8: 706036, 2021.
Article in English | MEDLINE | ID: mdl-35118081

ABSTRACT

BACKGROUND: It has been suggested that Helicobacter pylori (H. pylori) infection is associated with hypergastrinemia and proliferation of colorectal mucosa via direct stimulation, dysbiosis of the gut microbiome, and changes in the gut microbiome, all of which may lead to the formation of colorectal polyps. However, the consensus remains lacking regarding whether H. pylori infection is independently associated with colorectal polyps and whether the association differs according to histological type of colorectal polyps. To summarize the current evidence regarding the relationship between H. pylori infection and colorectal polyps, we conducted a meta-analysis of related observational studies according to the histological types of colorectal polyps. METHODS: Observational studies investigating the association between H. pylori infection and colorectal polyps using multivariate analyses were included by search of PubMed, Embase, and Web of Science. A random-effects model was adopted to combine the results. RESULTS: Seventeen studies that include 322,395 participants were analyzed. It was shown that H. pylori infection was independently associated with overall colorectal polyps (odds ratio [OR]: 1.67, 95% CI: 1.24-2.24, p < 0.001; I 2 = 73%). According to the histological type of colorectal polyps, H. pylori infection was independently associated with adenomatous polyps (APs; OR: 1.71, 95% CI: 1.47-1.99, p < 0.001; I 2 = 86%), advanced APs (OR: 2.06, 95% CI: 1.56-2.73, p < 0.001; I 2 = 0%), and hyperplastic polyps (HPs; OR: 1.54, 95% CI: 1.02-2.30, p = 0.04; I 2 = 78%). Evidence based on only one study showed that H. pylori infection was not associated with sessile serrated polyps (SSPs; OR: 1.00, 95% CI: 0.93-1.07, p = 0.99). CONCLUSIONS: Current evidence from case-control and cross-sectional studies suggested that H. pylori infection was independently associated with colorectal APs, advanced APs, and HPs, but not with SSPs. These findings suggested H. pylori infection may be a possible risk factor of colorectal polyp, which is important for the prevention of colorectal polyp in the adult population.

3.
J Clin Lab Anal ; 35(1): e23578, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32990355

ABSTRACT

BACKGROUND: Considering the boosting effect of glycolysis on tumor chemoresistance, this investigation aimed at exploring whether miR-488/PFKFB3 axis might reduce drug resistance of colorectal cancer (CRC) by affecting glycolysis, proliferation, migration, and invasion of CRC cells. METHOD: Totally, 288 CRC patients were divided into metastasis/recurrence group (n = 107) and non-metastasis/recurrence group (n = 181) according to their prognosis about 1 year after the chemotherapy, and their 3-year overall survival was also tracked. Besides, miR-488 expression was determined in peripheral blood of CRC patients and also in CRC cell lines (ie, W620, HT-29, Lovo, and HCT116). The targeted relationship between miR-488 and PFKFB3 was predicted by Targetscan software and confirmed by dual-luciferase reporter gene assay. Moreover, glycolysis and drug tolerance of CRC cells lines were assessed. RESULTS: MiR-488 expression was significantly decreased in metastatic/recurrent CRC patients than those without metastasis/recurrence (P < .05), and lowly expressed miR-488 was suggestive of unfavorable 3-year survival, large tumor size, poor differentiation, in-depth infiltration, and advanced Duke stage of CRC patients (P < .05). Besides, CRC cell lines transfected by miR-488 mimic demonstrated decreases in glucose uptake and lactate secretion, increases in oxaliplatin/5-Fu-sensistivity, as well as diminished capability of proliferating, invading, and migratory (P < .05), which were reversible by extra transfection of pcDNA3.1-PFKFB3 (ie, miR-488 mimic + pcDNA3.1-PFKFB3 group). Finally, the mRNA level of PFKFB3 was down-regulated by miR-488 mimic in CRC cell lines after being targeted by it (P < .05). CONCLUSION: The miR-488/PFKFB3 axis might clinically refine chemotherapeutic efficacy of CRC, given its modifying glycolysis and metastasis of CRC cells.


Subject(s)
Colorectal Neoplasms , Drug Resistance, Neoplasm/genetics , MicroRNAs , Phosphofructokinase-2 , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Glycolysis/genetics , Humans , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism
4.
Cell Biochem Funct ; 38(1): 47-57, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31710116

ABSTRACT

Colorectal cancer (CRC) is a common human malignancy that accounts for 600,000 deaths annually worldwide. Chrysophanol, a naturally occurring anthraquinone compound, exhibits anti-neoplastic effects in various cancer cells. The aim of this study was to explore the biological effects of chrysophanol on CRC cells, and determine the underlying mechanism. Chrysophanol inhibited proliferation of and promoted apoptosis in CRC cells by activating the intrinsic mitochondrial apoptotic pathway. In addition, chrysophanol also suppressed tumor growth in vivo and increased the percentage of apoptotic cells in tumor xenografts, without general toxicity. Proteomic iTRAQ analysis revealed decorin (DCN) as the major target of chrysophanol. DCN was upregulated in the tumor tissues following chrysophanol treatment, and ectopic DCN expression markedly augmented the pro-apoptotic effects of chrysophanol in CRC cells. In contrast, DCN knockdown significantly abrogated chrysophanol-induced apoptosis in CRC cells. Taken together, chrysophanol exerts anti-neoplastic effects in vitro and in vivo in CRC cells by modulating DCN, there by highlighting its therapeutic potential in CRC.


Subject(s)
Anthraquinones/pharmacology , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Decorin/antagonists & inhibitors , Animals , Anthraquinones/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Decorin/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , RNA, Small Interfering/pharmacology , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Anat Rec (Hoboken) ; 302(9): 1561-1570, 2019 09.
Article in English | MEDLINE | ID: mdl-30737895

ABSTRACT

Colorectal cancer (CRC) is a common human malignancy, accounting for 600,000 death cases annually worldwide. Chrysophanol is a naturally occurring anthraquinone compound and exhibits anti-neoplastic activities. This study aims to explore the biological effects of chrysophanol on CRC metastasis and the relevant underlying mechanism. Cell proliferation assay, wound scratch assay, and Transwell invasion assay were used to examine the effect of chrysophanol on proliferation, migration, and invasion of CRC cells. Hypoxia-inducible factor-1α (HIF-1α) shRNA was utilized to transfect CRC cells to examine the role of HIF-1α in chrysophanol suppression of hypoxia-induced epithelial to mesenchymal transition (EMT). The suppression effect of chrysophanol on hypoxia-induced EMT in vivo was also validated in xenograft tumor models. In the present study, our findings indicated that chrysophanol has the capability to suppress hypoxia-induced EMT in CRC in vitro and in vivo, and the possible mechanism involved is the inhibition of HIF-1α via modulating PI3k/Akt signaling pathway. Collectively, the results indicated that chrysophanol can be used as an EMT and cancer metastasis inhibitor in the treatment of CRC. Anat Rec, 302:1561-1570, 2019. © 2019 American Association for Anatomy.


Subject(s)
Anthraquinones/pharmacology , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Hypoxia/physiopathology , Mutagens/pharmacology , Animals , Apoptosis , Cell Movement , Cell Proliferation , Colorectal Neoplasms/drug therapy , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Cell Cycle ; 17(6): 728-738, 2018.
Article in English | MEDLINE | ID: mdl-29480061

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common causes of cancer-related death worldwide. In China, the situation is even worse as cancer incidence and mortality continue to increase rapidly. Although tremendous progress has been made toward HCC treatments, the benefits for liver cancer patients are still limited. Therefore, it is necessary to identify and develop novel therapeutic methods. Neuronally expressed developmentally downregulated 4 (NEDD4), an E3 ubiquitin ligase, plays a critical role in the development and progression of various types of human cancers. In our study, NEDD4 acts as an oncoprotein in both QGY7703 and SMMC7721 liver cancer cell lines. We found that depletion of NEDD4 by siRNA transfection led to inhibition of cell growth, invasion and migration, and promotion of apoptosis. In contrast, overexpression of NEDD4 via plasmid transfection resulted in facilitated cell proliferation, invasion and migration, and decreased apoptosis. Importantly, we observed that tumor suppressor LATS1, also a core component of Hippo pathway, was negatively regulated by NEDD4 in liver cancer cells. Our findings suggested that NEDD4 may be involved in the HCC progression via regulating LATS1 associated signaling pathway. Therefore, targeting NEDD4-LATS1 signaling could be a potential therapeutic option for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Liver Neoplasms/physiopathology , Nedd4 Ubiquitin Protein Ligases/metabolism , Apoptosis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Nedd4 Ubiquitin Protein Ligases/antagonists & inhibitors , Nedd4 Ubiquitin Protein Ligases/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism
7.
Oncol Lett ; 13(5): 3850-3858, 2017 May.
Article in English | MEDLINE | ID: mdl-28521485

ABSTRACT

Cyclooxygenase-2 (COX-2) and extracellular signal-regulated kinase 1/2 (ERK1/2) may serve as potential targets in various types of cancer; however, the roles of these proteins in gallbladder carcinoma (GBC) have not been reported previously. In the present study, the expression levels of COX-2 and phospho (p)-ERK1/2 in GBC were examined and the biological activities of celecoxib and PD184161 (specific inhibitors of COX-2 and p-ERK1/2, respectively) on the proliferation, cell cycle and apoptosis of the GBC-SD and NOZ human GBC cell lines were evaluated by a series of in vitro and in vivo studies. COX-2 and p-ERK1/2 protein expression levels were found to be significantly elevated in GBC tissues as well as in GBC-SD and NOZ cells. Treatments with celecoxib and PD184161 significantly inhibited GBC-SD and NOZ cell growth in a concentration-dependent manner, and their combination produced a synergistic inhibitory effect. In addition, celecoxib and PD184161 significantly inhibited tumor growth in xenograft nude mice. Celecoxib treatment led to G1 arrest via the upregulation of p21 and p27 expression in GBC-SD and NOZ cells, whereas PD184161 did not affect cell cycle distribution. The combination of celecoxib and PD184161 was able to promote cell apoptosis by triggering a collapse of mitochondrial membrane potential and activating caspase-3-mediated apoptosis. In conclusion, COX-2 and p-ERK1/2 protein may serve as potential targets for GBC chemotherapy, and the combination of celecoxib and PD184161 could significantly inhibit GBC cell growth, induce cell G1 arrest and trigger cell apoptosis of GBC cells.

8.
Oncotarget ; 6(29): 27805-15, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26314959

ABSTRACT

MicroRNAs (miRNAs) play a critical role in controlling tumor invasion and metastasis via regulating the expression of a variety of targets, which act as oncogenes or tumor suppressor genes. Abnormally expressed miR-625-3p has been observed in several types of human cancers. However, the molecular mechanisms of miR-625-3p-mediated tumorigenesis are largely elusive. Therefore, the aim of this study was to evaluate the biological function and molecular insight on miR-625-3p-induced oncogenesis in colorectal carcinoma (CRC). The effects of miR-625-3p in cell migration and invasion were analyzed by wound healing assay and transwell assay, respectively. In addition, the expression of miR-625-3p and its targets was detected in five human CRC cell lines. In the present study, we found that overexpression of miR-625-3p promoted migration and invasion in SW480 cells, whereas downregulation of miR-625-3p inhibited cell motility in SW620 cells. More importantly, we observed potential binding sites for miR-625-3p in the 3'-untranslated region of suppressor of cancer cell invasion (SCAI). Notably, we identified that overexpression of miR-625-3p inhibited the expression of SCAI, while depletion of miR-625-3p increased SCAI level, suggesting that SCAI could be a target of miR-625-3p. Additionally, we revealed that miR-625-3p exerts its oncogenic functions through regulation of SCAI/E-cadherin/MMP-9 pathways. Our findings indicate the pivotal role of miR-625-3p in invasion that warrants further exploration whether targeting miR-625-3p could be a promising approach for the treatment of CRC.


Subject(s)
Ataxin-1/biosynthesis , Cell Movement , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Blotting, Western , Cell Line, Tumor , Cell Movement/genetics , Colorectal Neoplasms/pathology , Humans , Neoplasm Invasiveness , Real-Time Polymerase Chain Reaction , Transfection
9.
Zhonghua Fu Chan Ke Za Zhi ; 38(9): 556-9, 2003 Sep.
Article in Chinese | MEDLINE | ID: mdl-14680612

ABSTRACT

OBJECTIVE: To investigate the effect of ceramide monohexoside (CMH) on resistance to cisplatin and apoptosis in ovarian cell line COC1/DDP, and to provide new ideals and clues to seek new effective methods for studying the mechanism and reversing the resistance in ovarian cell line as well. METHODS: COC1 cells and COC1/DDP cells (before and after the treatment of mifepristone) were collected and neutral glycosphingolipids (N-GSLs) of the cells was isolated and purified, changes of CMH content were analyzed by high performance thin layer chromatography (HPTLC). The COC1/DDP cells were divided into three groups, one treated by cisplatin, one treated by mifepristone, the other treated by cisplatin and mifepristone. The survival rate of cells in three groups were evaluated by the methyl thiazolyl tetrazolium (MTT) assay, DNA ladders were presented by DNA gel electrophoresis, the forms of cells were observed by transmission electron microscope (TEM). RESULTS: The levels of CMH were (37.1 +/- 3.3)% in COC1/DDP, higher than that in COC1 (14.1 +/- 1.4)% (P < 0.001). After treating by 1.25, 5 micro mol/L mifepristone, the CMH were (26.6 +/- 2.6)% (P < 0.05) and (17.5 +/- 0.7)% (P < 0.001), respectively. Mifepristone had no effect on the viability of COC1/DDP cell below a concentration of 5 micro mol/L. But when mifepristone of 1.25 or 5 micro mol/L combined with cisplatin at a concentration of 0.1, 0.25, 0.5, 1.25, 2.5 micro g/ml, the inhibition rate of COC1/DDP cell is higher than that of COC1/DDP cells only treated by cisplatin at the concentration of 0.1 to 2.5 micro g/ml (P < 0.001). The combined treatment elicited DNA fragmentation, however, neither cisplatin of 1.25 micro g/ml nor mifepristone of 5 micro mol/L alone could potentiate DNA fragmentation. After the combined treatment, the COC1/DDP cells produced apoptosis body. CONCLUSIONS: CMH is related with resistance to cisplantin in ovarian cell line COC1/DDP. When CMH of COC1/DDP cells was inhibited by mifepristone, the cells were sensitive to cisplatin and apoptosis was elicited.


Subject(s)
Cerebrosides/physiology , Cisplatin/pharmacology , Ovarian Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Cerebrosides/analysis , Cerebrosides/antagonists & inhibitors , DNA, Neoplasm/analysis , Drug Resistance, Neoplasm , Female , Humans , Ovarian Neoplasms/pathology , Ovarian Neoplasms/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...