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.
Transl Cancer Res ; 12(7): 1703-1714, 2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37588740

ABSTRACT

Background: Porcupine O-acyltransferase (PORCN), a membrane-bound O-acyltransferase, is crucial in Wnt ligand palmitoylation. However, the roles of PORCN in the development of hepatocellular carcinoma (HCC) remain unknown. Methods: Western blot, real-time quantitative polymerase chain reaction (RT-qPCR) assays, and The Cancer Genome Atlas (TCGA) database were used to study the expression and prognostic values of PORCN in patients with HCC. Following this, Cell Counting Kit-8 (CCK-8), wound-healing tests, Transwell assay, and a xenograft mouse model were employed to examine the effect of PORCN on HCC cells. Finally, the underlying molecular mechanisms involved in cell proliferation and migration caused by PORCN were identified. Results: The protein and messenger RNA (mRNA) levels of PORCN in HCC tissues were higher than those of adjacent normal tissues. The analysis of TCGA database indicated that patients with higher PORCN expression had a lower overall survival (OS) rate. Overexpression of PORCN could promote the proliferation and migration abilities of HCC cells both in vitro and in vivo. Gene set enrichment analysis (GSEA) showed that the effect of PORCN on the biological characteristics of HCC cells mainly centered on the Wnt-ß-catenin signaling pathway. Mechanically, immunofluorescence staining and subcellular protein fraction assays showed that PORCN could induce epithelial-mesenchymal transition (EMT) by promoting the translocation of ß-catenin from the cytoplasm to nucleus, ultimately promoting the progression of HCC. Conclusions: The findings of this study suggest that PORCN can promote HCC cell proliferation and migration by stimulating the Wnt-ß-catenin signaling pathway. Therefore, PORCN may be a promising therapeutic target for HCC.

2.
Sci Total Environ ; 846: 157414, 2022 Nov 10.
Article in English | MEDLINE | ID: mdl-35850325

ABSTRACT

Fe-loaded biochar (FeBC) has been considered for Sb(III) adsorption, but the effects of oxygen (O2) on the adsorption need further investigation. Liquid-/solid-phase analyses were conducted to investigate the role of O2 in the Sb(III) adsorption by FeBC. The adsorption was best described by the pseudo-second-order (PSO) model for kinetic results and by the Langmuir model for thermodynamic results. More than 96.8 % of Sb(III) was adsorbed by FeBC, and available O2 increased the liquid-phase Sb(III) oxidation efficiency by 2.1-7.5 times. The peak changes at ~1640 and 3450 cm-1 in FTIR spectra indicated the occurrence of inner-sphere complexation between Sb(III)/Sb(V) and hydroxyl (-OH)/carboxyl (-COOH) groups in FeBC under aerobic and anaerobic conditions. Fe/Sb X-ray absorption spectroscopy (XAS) analysis results showed aqueous Sb(III) complexed to the edge-sharing Fe(III)-O-Fe(III) in FeBC. Regardless of whether O2 was available or not, solid-phase edge-sharing Fe(III)-O-Sb(V) complexes (~3.05 Å), which had lower toxicity and migration ability than aqueous Sb(III), formed through a ligand-to-metal charge-transfer (LMCT) process. More than 91 % of adsorbed Sb(III) was oxidized to edge-sharing Fe(III)-O-Sb(V) complexes in 3 h. Additionally, the Sb(V) from liquid-phase oxidation could also directly complex to the Fe(III)-O-Fe(III) and form edge-sharing Fe(III)-O-Sb(V) complexes. These results provide evidence to inform further FeBC application for the Sb-contaminated water treatment.


Subject(s)
Antimony , Ferric Compounds , Adsorption , Antimony/chemistry , Charcoal , Ferric Compounds/chemistry , Oxygen , X-Ray Absorption Spectroscopy
3.
Int J Gen Med ; 15: 885-895, 2022.
Article in English | MEDLINE | ID: mdl-35115815

ABSTRACT

BACKGROUND: As hepatocellular carcinoma (HCC) having the second-highest mortality rate globally, the early diagnosis and prognosis of HCC have always been the focus of various studies. Although PSME4 has been reported to be closely related to several malignancies, its role in HCC remains unclear. MATERIALS AND METHODS: The TCGA-LIHC database and HCC tissues were used to explore the expression of PSME4 in HCC. Gene set enrichment analysis (GSEA) was used to forecast the biological behavior of HCC cells that PSME4 might be involved in regulation. In addition, CCK-8, colony formation and flow cytometry assays were used to explore the effect of PSME4 on HCC cells. Furthermore, the underlying PSME4-related signaling pathways in HCC were further confirmed using GSEA. RESULTS: We found that the expression of PSME4 in HCC tissues was significantly higher than that in adjacent normal tissues, and patients with high PSME4 expression have a poor prognosis. CCK-8, colony formation and flow cytometry assays shown that knockdown of PSME4 inhibits HCC cell proliferation of HCC cells, promotes cell apoptosis and moves the cell cycle away from the S phase. Mechanistically, PSME4 may promote the development of HCC through mTOR signaling pathway. CONCLUSION: The high expression of PSME4 in HCC promotes the proliferation of HCC cells via the mTOR signalling pathway. Therefore, PSME4 is an emerging tumour marker for the early diagnosis and prognosis of HCC.

4.
Bioengineered ; 12(2): 12598-12611, 2021 12.
Article in English | MEDLINE | ID: mdl-34935584

ABSTRACT

Colon adenocarcinoma (COAD), having high malignancy and poor prognosis, is the main pathological type of colon cancer. Previous studies show that Keratin 17 (KRT17) plays an important role in the development of many malignant tumors. However, its role and the molecular mechanism underlying COAD remain unclear. Using TCGA and ONCOMINE databases, as well as immunohistochemistry, we found that the expression of KRT17 was higher in COAD tissues as compared to that in the adjacent normal tissues. Cell- and animal-based experiments showed that overexpression of KRT17 promoted the invasion and metastasis of colon cancer cells while knocking down KRT17 reversed these processes both in vitro and in vivo. In addition, we also showed that KRT17 promoted the formation of new blood vessels. Mechanistically, KRT17 could regulate the WNT/ß-catenin signaling pathway, and APC may be involved in this process by interacting with KRT17. In summary, these findings suggested that high expression of KRT17 could promote cell metastasis and angiogenesis of colon cancer cells by regulating the WNT/ß-catenin signaling pathway. Thus, KRT17 could be a potential therapeutic target for COAD treatment.


Subject(s)
Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Keratin-17/genetics , Neovascularization, Pathologic/genetics , Up-Regulation , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Cell Line, Tumor , Chickens , Colonic Neoplasms/genetics , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Keratin-17/metabolism , Male , Mice, Inbred C57BL , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Up-Regulation/genetics , Wnt Signaling Pathway/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...