Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
BMC Gastroenterol ; 23(1): 343, 2023 Oct 03.
Article in English | MEDLINE | ID: mdl-37789294

ABSTRACT

BACKGROUND: The routine establishment of a diverting stoma (DS) remains controversial in every patient undergoing Dixon operation. We aimed to establish a model for the risk assessment of rectal anastomotic leak (RAREAL) after Dixon in non-emergency patients with rectal cancer, using routinely available variables, by which surgeons could individualize their approach to DS. METHODS: 323 patients who underwent Dixon operation for rectal cancer from January 2015 to December 2018 were taken as the model group for retrospective study. Univariable and multivariable logistic regression analysis was used to determine the independent risk factors associated with anastomotic leakage. We constructed the RAREAL model. 150 patients who underwent Dixon operation due to rectal cancer from January 2019 to December 2020 were collected according to the uniform criteria as a validation group to validate the RAREAL model. RESULTS: In the model group, multivariable analysis identified the following variables as independent risk factors for AL: HbA1c (odds ratio (OR) = 4.107; P = 0.044), Left colic artery (LCA) non preservation (OR = 4.360; P = 0.026), Tumor distance from the anal margin (TD) (OR = 6.373; P = 0.002). In the model group, the area under the curve (AUC) of the receiver operating characteristic (ROC) for evaluating AL with RAREAL was 0.733, and when RAREAL score = 2.5, its sensitivity, specificity and Youden index were 0.385, 0.973, 0.358, respectively. The AUC was 0.722 in the validation group and its sensitivity and specificity were 0.333 and 0.985, respectively, when RAREAL score = 2.5. CONCLUSION: The RAREAL score can be used to assess the risk of AL after Dixon operation for rectal cancer, and prophylactic DS should be proactively done when the score is greater than 2.5.


Subject(s)
Gastrointestinal Diseases , Rectal Neoplasms , Humans , Anastomotic Leak/etiology , Anastomosis, Surgical/adverse effects , Retrospective Studies , Rectal Neoplasms/surgery , Rectal Neoplasms/complications , Risk Assessment , Risk Factors , Gastrointestinal Diseases/etiology
2.
J BUON ; 25(3): 1390-1394, 2020.
Article in English | MEDLINE | ID: mdl-32862581

ABSTRACT

PURPOSE: Liver cancer stem cells are associated with tumor progression, metastasis, and resistance to chemotherapy. Therefore, it is important to understand the proteins that support the tumor microenvironment. The suppression of ZEB2 results from inactivation of the Wnt/ß catenin pathway. Like RBM38, it suppresses tumor outgrowth and helps increase the survival of cancer patients. However, no studies have examined the direct roles of ZEB2 and RBM38 in the tumor microenvironment. METHODS: We developed an early/advanced stage liver cancer mouse model using CD133+ cell injection that mimics liver cancer in all ways. Histology, Western blotting, and immunohistochemistry analyses were used to examine cancer progression. RESULTS: Histologically, the early liver cancer showed microfoci structures; the advanced cancer showed distinct morphological changes with enlarged nucleoli and cell clumping. Immunohistochemical and Western blotting analyses of CD133 and ZEB2 proteins showed similar upregulated expression as the tumor progressed. However, RBM38 expression increased dramatically in early liver cancer but was downregulated in advanced liver cancer. CONCLUSIONS: ZEB2 favors a tumor microenvironment that supports liver cancer stem cell proliferation, while RBM38 expression negatively affects the tumor microenvironment and restricts liver cancer stem cell proliferation.


Subject(s)
Cell Proliferation/genetics , Liver Neoplasms/genetics , Neoplastic Stem Cells/pathology , RNA-Binding Proteins/genetics , Zinc Finger E-box Binding Homeobox 2/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Disease Models, Animal , Down-Regulation/genetics , Male , Mice , Mice, Inbred BALB C , Tumor Microenvironment/genetics , Up-Regulation/genetics , Wnt Signaling Pathway/genetics
3.
Cancer Manag Res ; 12: 12185-12195, 2020.
Article in English | MEDLINE | ID: mdl-33273855

ABSTRACT

PURPOSE: LACTB, regulated by a variety of microRNAs (miRNAs), is proven to be a tumor suppressor. However, there are few reports that LACTB in colon cancer cells is regulated by miRNA. Therefore, the aim of this study was to explore the miRNAs that regulate LACTB in colon cancer. PATIENTS AND METHODS: Data from TCGA were analyzed in starBase and GEPIA2, and Western blot and quantitative PCR (qPCR) were used to detect the expression of LACTB in colon cancer cell lines. MiRNAs targeting LACTB were predicted by MicroT-CDS, starBase, miRDB, mirDIP, and DIANA. The relationship between LACTB and miRNA was explored by dual-luciferase assay. MTT, propidium iodide (PI), Western blot, Annexin V-FITC/PI Kit, qPCR and transwell assay were used to detect the changes in cell proliferation, cell cycle, autophagy, apoptosis, epithelial-to-mesenchymal transition (EMT), cell migration, and invasiveness in colon cancer cells that overexpressed miR-1276 and/or LACTB. RESULTS: The results showed that the LACTB mRNA level was lower and the miR-1276 level was higher in colon cancer than in normal tissue. MiR-1276 inhibited the expression of LACTB. Furthermore, overexpression of miR-1276 in colon cancer cells increased proliferation, migration, invasiveness and EMT, and decreased autophagy and apoptosis. Supplementing LACTB suppressed these effects of miR-1276. CONCLUSION: In conclusion, miR-1276, which may be a potential therapy for colon cancer, inhibits cell growth and promotes apoptosis by targeting LACTB in colon cancer cells.

4.
Integr Cancer Ther ; 19: 1534735420972477, 2020.
Article in English | MEDLINE | ID: mdl-33289438

ABSTRACT

Protosappanin B (PSB) is a key active component of Lignum Sappan extract. Although the antiproliferative effects of Lignum Sappan extract have been demonstrated in various cancer cells, relatively little is known about the effects of PSB on tumor progression. The aim of this study was to explore the anti-tumor effects of PSB on human colon cancer cells by regulation of intracellular signaling pathways and Golgi phosphoprotein 3 (GOLPH3) expression in vitro and in vivo. Our results showed that PSB effectively inhibited the viability and migration of SW620 cells and induced apoptosis, but had poor effect on HCT116 cells. Furthermore, PSB significantly reduced the expression of p-AKT, p-p70S6K, ß-catenin, and p-ERK1/2 proteins in SW620 cells, and this effect was reversed by the corresponding signaling pathway agonists. Interestingly, PSB could also suppress GOLPH3 expression of SW620 cells in a concentration-dependent manner, but SW620 cells transfected with lentiviral vectors overexpressing GOLPH3 can effectively resist the cytotoxic activity of PSB in vitro. The xenograft experiment of SW620 cells with LV-GOLPH3 confirmed that PSB distinctly inhibited the tumor growth via suppressing GOLPH3 expression. Collectively, these findings clarified a new anti-cancer mechanism of PSB through inhibition of GOLPH3 expression and intracellular signaling pathways in colon cancer cells. PSB may be a potential new drug for colon cancer.


Subject(s)
Colonic Neoplasms , Oxocins , Cell Line, Tumor , Colonic Neoplasms/drug therapy , HCT116 Cells , Humans , Membrane Proteins , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL