Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Cancer Res Clin Oncol ; 149(11): 8495-8505, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37095412

ABSTRACT

BACKGROUND: Gastric cancer (GC) remains a global challenge due to its high morbidity and mortality rates especially in Asia as well as poor response to treatment. As a member of the adhesion protein family and transmembrane glycoprotein, EpCAM expressed excessively in cancer cells including GC cells. The database assay showed that EpCAM is excessively expressed and easily mutated in cancers, especially in early stage of GC. METHODS: To explore the roles EpCAM plays in oncogenesis and progression of GC, the expression of EpCAM was deleted in GC cells with CRISPR/Cas9 method, and then the changes of cell proliferation, apoptosis, motility and motility associated microstructures in EpCAM-deleted GC cells (EpCAM-/-SGC7901) were detected to evaluate the rules EpCAM played. RESULTS: The results showed that EpCAM deletion caused cell proliferation, motility and the development of motility-relevant microstructures inhibited significantly, apoptotic trend and contact inhibition enhanced in EpCAM-deleted GC cells. The results of western blot suggested that EpCAM modulates the expression of epithelial/endothelial mesenchymal transition (EMT) correlated genes. All results as above indicated that EpCAM plays important roles to enhance the oncogenesis, malignancy and progression as a GC enhancer. CONCLUSIONS: Combining our results and published data together, the interaction of EpCAM with other proteins was also discussed and concluded in the discussion. Our results support that EpCAM can be considered as a novel target for the diagnosis and therapy of GC in future.


Subject(s)
Stomach Neoplasms , Humans , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Stomach Neoplasms/pathology , Proteins/genetics , Carcinogenesis/genetics , Asia , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Cell Proliferation , Cell Movement/genetics , Gene Expression Regulation, Neoplastic
2.
Biotechnol Lett ; 44(8): 951-960, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35771408

ABSTRACT

OBJECTIVES: Ovarian cancer is one of the most fatal gynecological malignancies. It is emergently needed to select a novel molecular fragment as a targeting element for the future development of molecular imaging diagnosis and targeting chemotherapy to ovarian cancer. RESULTS: After five rounds of biopanning, a total of 44 positive phage clones were selected from final phage displayed peptide library. Nine consensus sequences were found based on the assay of sequencing results, then one clone of each consensus group was characterized and identified further by immunofluorescence assay. The result showed the phage clone R20 presents best targeting capacity. Then we synthesized peptide (OSP2) clone R20 displayed, it was characterized with high specificity and sensitivity binding to human ovarian cancer by a tissue chip assay. The target of OSP2 was predicted and docked as human carbonic anhydrase XII (CA12), an important protein usually deregulated in cancer. CONCLUSIONS: Taken together, OSP2 and its target indicate a novel investigation way in future to develop novel agent or drug delivery formulation for molecular imaging diagnosis and targeting chemotherapy of ovarian cancer.


Subject(s)
Bacteriophages , Ovarian Neoplasms , Bacteriophages/metabolism , Cell Line, Tumor , Female , Humans , Peptide Library , Peptides/chemistry , Protein Binding
3.
Int J Mol Sci ; 22(19)2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34639214

ABSTRACT

Lymphoid enhancer-binding factor 1 (LEF1) is a key transcription factor mediating the Wnt signaling pathway. LEF1 is a regulator that is closely associated with tumor malignancy and is usually upregulated in cancers, including colonic adenocarcinoma. The underlying molecular mechanisms of LEF1 regulation for colonic adenocarcinoma progression remain unknown. To explore it, the LEF1 expression in caco2 cells was inhibited using an shRNA approach. The results showed that downregulation of LEF1 inhibited the malignancy and motility associated microstructures, such as polymerization of F-actin, ß-tubulin, and Lamin B1 in caco2 cells. LEF1 inhibition suppressed the expression of epithelial/endothelial-mesenchymal transition (EMT) relevant genes. Overall, the current results demonstrated that LEF1 plays a pivotal role in maintaining the malignancy of colonic adenocarcinoma by remodeling motility correlated microstructures and suppressing the expression of EMT-relevant genes. Our study provided evidence of the roles LEF1 played in colonic adenocarcinoma progression, and suggest LEF1 as a potential target for colonic adenocarcinoma therapy.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , Cell Movement , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Lymphoid Enhancer-Binding Factor 1/metabolism , Actins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Humans , Lymphoid Enhancer-Binding Factor 1/genetics , Pseudopodia/metabolism , Tubulin/metabolism , Tumor Cells, Cultured
4.
Int J Pharm ; 599: 120446, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33675923

ABSTRACT

Gastric cancer (GC) presents a challenge for conventional therapeutics due to low targeting specificity and subsequent elicitation of multiple drug resistance (MDR). As an essential enzyme for DNA repair, apurinic/apyrimidinic endodeoxyribonuclease 1 (APEX1) exhibits multiple functions to affect cancer malignancy and is excessively expressed in GC. However, the roles APEX1 and its inhibitor miR-27a-5p play in modulating GC progression and MDR development remains unclear. Here, we verified APEX1 as a target of miR-27a-5p and subsequently established the APEX1-deleted SGC-7901 cell line by CRISPR/Cas9 editing. The roles of the APEX1/miR-27a-5p axis in GC progression, metastasis and doxorubicin (DOX) resistance were explored by the targeted chemotherapy facilitated by a GC-specific peptide (GP5) functionalized liposomal drug delivery formulation (GP5/Lipo/DOX/miR-27a-5p). The results showed that APEX1 deletion distinctly attenuated cell growth and metastatic properties in GC, and also sensitized GC cells to DOX. Notably, miR-27a-5p was validated as a suppressor of APEX1-dependent GC development and DOX resistance by a RAS/MEK/FOS and PTEN/AKT/SMAD2 pathway-dependent manner. The altered expression of epithelial-mesenchymal transition (EMT) signatures and signal pathway proteins in the APEX1-deleted cells implied that APEX1 potentially enhances DOX resistance of GC cells by altering the regulation of MAPK and AKT pathways, leading to compromised efficacy of chemotherapy or by initiating additional DNA damage response pathways. Taken together, these findings revealed that as a novel therapeutic target, APEX1/miR-27a-5p axis plays essential roles in modulating the GC development and MDR, and the GC targeted drug delivery formulation presents a strategic reference for the future designation of chemotherapeutics study.


Subject(s)
MicroRNAs , Stomach Neoplasms , Cell Line, Tumor , DNA-(Apurinic or Apyrimidinic Site) Lyase , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Signal Transduction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...