Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 5 de 5
1.
Front Oncol ; 12: 802009, 2022.
Article En | MEDLINE | ID: mdl-35251974

BACKGROUND: This study aims to investigate the effects of ω-3, ω-6 polyunsaturated fatty acids (PUFAs), and their middle metabolites prostaglandin (PGE)2 and PGE3 on proliferation, invasion, and angiogenesis formation of gastric cancer cells and to explore associated mechanism. METHODS: RT-PCR and ELISA were used to detect the expression of cyclooxygenase (COX)-1 and COX-2 in gastric cancer cell lines. The effect of ω-3, ω-6, PGE2, and PGE3 on the proliferation, invasion, and angiogenesis of gastric cancer cells were measured by cell proliferation, invasion, and angiogenesis assay in vitro. COX-2 small interfering RNA (siRNA) was transfected into gastric cancer cells, and the expression of COX-2 protein was detected by Western blot. COX-2 gene silencing influencing proliferation, invasion, and angiogenesis potential of gastric cancer cells was detected by WST-1, transwell chamber, and angiogenesis assay, respectively. RESULTS: COX-2 was only expressed in MKN74 and MKN45 cells. In gastric cancer cell lines with positive COX-2 expression, ω-6 and PGE2 could significantly enhance the proliferation, invasion, and angiogenesis of gastric cancer cells, and after transfection with COX-2 siRNA, the effects of ω-6 and PGE2 on enhancing the proliferation, invasion, and angiogenesis of gastric cancer cells were significantly attenuated; ω-3 and PEG3 could inhibit the proliferation, invasion, and angiogenesis of gastric cancer cells. In gastric cancer cell lines with negative COX-2 expression, ω-6 and PGE2 had no significant effect on the proliferation, invasion, and angiogenesis of gastric cancer; ω-3 and PGE3 could significantly inhibit the proliferation, invasion, and angiogenesis of gastric cancer. CONCLUSION: ω-6 PUFAs reinforce the metastatic potential of gastric cancer cells via COX-2/PGE2; ω-3 PUFAs inhibit the metastatic potential of gastric cancer via COX-1/PGE3 signaling axis.

2.
Cell Commun Signal ; 19(1): 122, 2021 12 20.
Article En | MEDLINE | ID: mdl-34930323

BACKGROUND: The aim of this study was to investigate the co-operative role of CXCR4/CXCL12 axis and IL-1Ra in metastatic processes mechanism by interactions between colorectal cancer cells and stromal cells in their microenvironment. METHODS: Expression of IL-1α, interleukin-1 receptor type I (IL-1 RI), CXCL12 and CXCR4 mRNA and proteins were determined by RT-PCR and Western blot. The effect of secreted level of CXCL12 by IL-1Ra on fibroblasts was measured by ELISA. CXCL12 regulate metastatic potential of colorectal cancer was evaluated by proliferation, invasion and angiogenesis assays, respectively, in which invasion and angiogenesis assays used an in vitro system consisting of co-cultured colorectal cells and stromal cells. RESULTS: IL-1α was expressed in high liver metastatic colorectal cancer cell lines (HT-29 and WiDr). The colorectal cancer cell-derived IL-1α and rIL-1α significantly promoted CXCL12 expression by fibroblasts, and this enhancing effect can be significantly inhibited by IL-1Ra (P < 0.01). CXCL12 not only enhanced the migration and proliferation of human umbilical vein endothelial cells, but also significantly enhanced angiogenesis (P < 0.01). Furthermore, the high liver-metastatic colorectal cancer cell line (HT-29), which secretes IL-1α, significantly enhanced angiogenesis compared to the low liver-metastatic cell line (CaCo-2), which does not produce IL-1α (P < 0.01). On the contrary, IL-1Ra can significantly inhibit migration, proliferation and angiogenesis (P < 0.01). CONCLUSION: Autocrine IL-1α and paracrine CXCL12 co-enhances the metastatic potential of colorectal cancer cells; IL-1Ra can inhibit the metastatic potential of colorectal cancer cells via decrease IL-1α/CXCR4/CXCL12 signaling pathways. Video Abstract.


Interleukin 1 Receptor Antagonist Protein
3.
Eur J Cancer Prev ; 30(6): 442-447, 2021 11 01.
Article En | MEDLINE | ID: mdl-34596103

The purpose of paper is to investigate the depression and anxiety as well as independent influential factors between patients who underwent Da Vinci robot-assisted radical gastrectomy and radical gastrectomy. This study is a partially randomized patient preference trial. A total of 98 patients with gastric cancer were divided into the Da Vinci robot-assisted radical gastrectomy group (46 patients, observation group) and open radical gastrectomy group (52 patients, control group). They were also postoperatively and preoperatively measured with Self-Rating Depression Scale (SDS) and Self-Rating Anxiety Scale (SAS). The postoperative and preoperative data of each group were compared. The postoperative and preoperative standard scores of SDS and SAS in patients with the observation group were NS differences (P > 0.05). In the conventional control group, the postoperative mean scores of SDS and SAS were significantly higher than those in the preoperative mean scores and the postoperative mean scores of the observation group, respectively (P < 0.01). The multivariate logistic regression analysis indicated that the independent influential factors of depression and anxiety in patients with radical gastrectomy included tumor, node, metastases stage, pain grading, other postoperative complications and postoperative insomnia (P < 0.05). Robot-assisted radical gastrectomy for gastric carcinoma is conductive to relieving patients' anxiety and depression and improving their quality of life due to the advantages of relatively low incidence of pain, reduced complications and relatively good sleep.


Robotics , Stomach Neoplasms , Anxiety/diagnosis , Anxiety/epidemiology , Anxiety/etiology , Depression/diagnosis , Depression/epidemiology , Depression/etiology , Gastrectomy/adverse effects , Humans , Pain/surgery , Quality of Life , Retrospective Studies , Stomach Neoplasms/surgery , Treatment Outcome
4.
Drug Deliv ; 28(1): 1204-1213, 2021 Dec.
Article En | MEDLINE | ID: mdl-34142633

Gastric cancer (GC) remains a major public health problem. Ursolic acid (UA) is reported to be effective in inhibiting GC; however, its low solubility and poor biocompatibility have greatly hindered its clinical application. Herein, an innovative reactive oxygen species (ROS)-sensitive UA dimeric prodrug is developed by coupling two UA molecules via a ROS-cleavable linkage, which can self-assemble into stable nanoparticles in the presence of surfactant. This new UA-based delivery system comprises the following major components: (I) dimeric prodrug inner core that can achieve high drug-loading (55%, w/w) and undergo rapid and selective conversion into intact drug molecules in response to ROS; (II) a polyethylene glycol (PEG) shell to improve colloid stability and extend blood circulation, and (III) surface-modified internalizing RGD (iRGD) to increase tumor targeting. Enhancement of the antitumor effect of this delivery system was demonstrated against GC tumors in vitro and in vivo. This novel approach offers the potential for clinical applications of UA.


Nanoparticles/chemistry , Prodrugs/pharmacology , Reactive Oxygen Species/metabolism , Stomach Neoplasms/drug therapy , Triterpenes/pharmacology , Animals , Cell Line, Tumor , Cell Survival , Chemistry, Pharmaceutical , Drug Carriers/chemistry , Drug Liberation , Humans , Hydrogen-Ion Concentration , Male , Mice , Mice, Inbred BALB C , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Random Allocation , Triterpenes/administration & dosage , Triterpenes/pharmacokinetics , Xenograft Model Antitumor Assays , Ursolic Acid
5.
Eur J Cancer Prev ; 30(3): 220-231, 2021 05 01.
Article En | MEDLINE | ID: mdl-32701605

To better explore the underlying mechanism of liver metastatic formation by placenta-specific protein 1 (PLAC1) in human colorectal cancer, we investigated the proliferation, invasion and angiogenic capabilities of human colorectal cancer cells with different liver metastatic potentials as well as the mechanism of action of PLAC1 in the metastatic process. The expression of PLAC1 was detected by reverse transcriptase PCR, western blot, and real-time PCR. The effect of PLAC1 on metastatic potential was determined by proliferation, invasion, and angiogenesis assays, including an in-vitro coculture system consisting of cancer cells and vascular endothelial cells that were used to detect the relationship between cancer cells and angiogenesis. In addition, we also determined PLAC1 downstream targets that preferentially contribute to the metastatic process. PLAC1 was expressed in HT-29, WiDr, and CaCo-2 colorectal cancer cells but not in Colo320 colorectal cancer cells. PLAC1 not only enhanced significantly the proliferation of CoLo320 and human umbilical vein endothelial cells (HUVECs) but also promoted the invasion of CoLo320 cells. The angiogenesis of HUVECs was enhanced by PLAC1 in a dose-dependent manner. In cocultured systems, angiogenesis was significantly increased by coculture with HT-29 cells. In addition, PLAC1 could promote angiogenesis in coculture with HT-29 cells. Furthermore, PLAC1-enhanced metastatic potential of colorectal cancer cells was dependent on the activation of the PI3K/Akt/NF-κB pathway. The activation of PI3K/Akt/NF-κB signaling by PLAC1 may be critical for metastasis of colorectal cancer cells. According to our results, we suggest that modification of PLAC1 function might be a promising new therapeutic approach to inhibit the aggressive spread of colorectal cancer.


Colorectal Neoplasms , Pregnancy Proteins , Caco-2 Cells , Cell Movement/physiology , Cell Proliferation , Colorectal Neoplasms/genetics , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Liver/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases , Placenta/metabolism , Pregnancy , Pregnancy Proteins/metabolism , Pregnancy Proteins/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/pharmacology , Signal Transduction
...