Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
2.
Oncogene ; 40(38): 5764-5779, 2021 09.
Article in English | MEDLINE | ID: mdl-34341514

ABSTRACT

Anoikis is a type of programmed cell death induced by loss of anchorage to the extracellular matrix (ECM). Anoikis resistance (AR) is crucial for the survival of metastatic cancer cells in blood, lymphatic circulation and distant organs. Compared to ordinary cancer cells, anoikis resistant cancer cells undergo various cellular and molecular alterations, probably characterizing the cells with unique features not limited to anoikis resistance. However, the molecular mechanisms connecting anoikis resistance to other metastatic properties are still poorly understood. Here, the biological interaction between anoikis resistance and angiogenesis as well as their involvement into peritoneal metastasis of gastric cancer (GC) were investigated in vitro and in vivo. The prognostic value of key components involved in this interaction was evaluated in the GC cohort. Compared to ordinary GC cells, GCAR cells exhibited stronger metastatic and pro-angiogenic traits corresponding to elevated PDGFB secretion. Mechanistically, transcription factor C/EBPß facilitated PDGFB transcription by directly binding to and interacting with PDGFB promoter elements, subsequently increasing PDGFB secretion. Secreted PDGFB promoted the survival of detached GC cells through a C/EBPß-dependent self-feedback loop. Moreover, secreted PDGFB promoted angiogenesis in metastases via activation of the MAPK/ERK signaling pathway in vascular endothelial cells. Both C/EBPß activation level and PDGFB expression were significantly elevated in GC and correlated with metastatic progression and poor prognosis of patients with GC. Overall, interaction between GCAR cells and vascular endothelial cells promotes angiogenesis and peritoneal metastasis of GC based on C/EBPß-mediated PDGFB autocrine and paracrine signaling. C/EBPß-PDGFB-PDGFRß-MAPK axis promises to be potential prognostic biomarkers and therapeutic targets for peritoneal metastasis of GC.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/secondary , Proto-Oncogene Proteins c-sis/genetics , Stomach Neoplasms/pathology , Animals , Anoikis , Autocrine Communication , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Transplantation , Paracrine Communication , Peritoneal Neoplasms/blood supply , Peritoneal Neoplasms/metabolism , Prognosis , Promoter Regions, Genetic , Proto-Oncogene Proteins c-sis/metabolism , Stomach Neoplasms/blood supply , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Up-Regulation
3.
Biomed Pharmacother ; 126: 109941, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32169757

ABSTRACT

BACKGROUND: Gastric cancer (GC) has been regarded as a kind of the most common cancers in gastrointestinal malignant tumors. Circular RNA (circRNA) is a newly discovered category of non-coding RNAs and plays a significant role in the initiation or development of human cancers. Nevertheless, the role of circPIP5K1A in GC remains unclear. METHODS: The relative expression level and the circular structure of circPIP5K1A were confirmedby RT-qPCR. The biological function of circPIP5K1A in GC was evaluated by colony formation, transwell and western blot assays. The binding capacity between miR-671-5p and circPIP5K1A (or KRT80) was assessed by luciferase reporter and Ago2-RIP assays. Protein levels of PI3K/AKT pathway were measured by western blot assay. RESULTS: CircPIP5K1A was up-regulated in GC tissues and cells with a circular structure. Functionally, circPIP5K1A silence limited cell proliferation, invasion, migration and EMT process. Mechanistically, circPIP5K1A directly interacted with miR-671-5p to modulate KRT80 expression. Either miR-671-5p inhibitor or KRT80 overexpression could offset the inhibitory effect of circPIP5K1A depletion on GC development. Besides, circPIP5K1A played its oncogenic role in GC through regulating PI3K/AKT pathway. At last, circPIP5K1A promoted GC tumor growth in vivo. CONCLUSIONS: CircPIP5K1A/miR-671-5p/KRT80 axis contributes to GC progression through PI3K/AKT pathway, implying this axis may be a potential therapeutic target for the treatment of GC patients.


Subject(s)
Keratins, Type II/metabolism , MicroRNAs/metabolism , RNA, Circular/metabolism , Stomach Neoplasms/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/physiology , Humans , Keratins, Type II/genetics , MicroRNAs/genetics , RNA, Circular/genetics , Stomach Neoplasms/genetics , Up-Regulation
4.
Eur J Pharmacol ; 852: 189-197, 2019 Jun 05.
Article in English | MEDLINE | ID: mdl-30876979

ABSTRACT

The bromodomain and extra-terminal domain (BET) protein BRD4 is emerging as a potential target for cancer therapy. However, BRD4 roles in regulating the stemness of gastric cancer cells are unclear. Here, we demonstrated that BRD4 expression was significantly increased in gastric cancer tissues, cell spheroids, and BRD4 knockdown attenuated the stemness of gastric cancer cells characterized as the decrease of stemness markers expression, capacity of cells spheroids formation and ALDH1 activity. Importantly, BRD4 expression was negatively correlated with overall survival, first progression survival and post progression survival of gastric cancer patients. Mechanistic investigations revealed that miR-216a-3p was the most remarkably upregulated miRNA in response to BRD4 knockdown and Wnt/ß-catenin signaling was necessary for BRD4-mediated promotion on the stemness of gastric cancer cells. Additionally, BRD4 directly bound to the promoter and promoted the methylation level of MIR216A promoter, thus decreasing miR-216a-3p level. Notably, Wnt3a was identified as the direct target of miR-216a-3p in gastric cancer cells. Therefore, our results defined a BRD4/miR-216a-3p/Wnt/ß-catenin pathway in regulating the stemness of gastric cancer cells.


Subject(s)
MicroRNAs/genetics , Neoplastic Stem Cells/pathology , Nuclear Proteins/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcription Factors/metabolism , Wnt Signaling Pathway/genetics , Base Sequence , Cell Cycle Proteins , Cell Line, Tumor , Disease Progression , Gene Knockdown Techniques , Humans , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
5.
Biomed Pharmacother ; 108: 338-346, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30227327

ABSTRACT

This work aims to explore the roles and mechanisms of long non coding RNA (lncRNA) THOR in regulating the stemness of gastric cancer cells. RNA-sequencing combined with quantitative real-time PCR (qRT-PCR) indicated that lncRNA THOR level was significantly upregulated in gastric cancer tissues compared with that in normal adjacent tissues. Knockdown of THOR attenuated the stemnness of gastric cancer cells, evident by the decrease of stemness markers expression and capacity of cells spheroid formation. Further RNA-sequencing combined with qRT-PCR and western blot analysis demonstrated that expression of transcriptional factor SOX9 was remarkably decreased in gastric cancer cells with THOR stable knockdown. Additionally, RNA immunoprecipitation (RIP) combined with luciferase reporter assay revealed that THOR directly bound to SOX9 3' untranslated region (3'UTR), but not its 5'UTR or coding area. Notably, overexpression of SOX9 rescued THOR knockdown-mediated inhibition on the stemness of gastric cancer cells. Thus, our results suggest that THOR could potentiate the stemness of gastric cancer cells via directly binding to SOX9 3'UTR.


Subject(s)
Neoplastic Stem Cells/pathology , RNA Stability/genetics , RNA, Long Noncoding/metabolism , SOX9 Transcription Factor/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gene Knockdown Techniques , Humans , Neoplastic Stem Cells/metabolism , RNA, Long Noncoding/genetics , SOX9 Transcription Factor/metabolism
6.
Int J Nanomedicine ; 7: 1723-35, 2012.
Article in English | MEDLINE | ID: mdl-22619523

ABSTRACT

A novel formulation containing polyvinylpyrrolidone (PVP) K(30)-coated norcantharidin (NCTD) chitosan nanoparticles (PVP-NCTD-NPs) was prepared by ionic gelation between chitosan and sodium tripolyphosphate. The average particle size of the PVP-NCTD-NPs produced was 140.03 ± 6.23 nm; entrapment efficiency was 56.33% ± 1.41%; and drug-loading efficiency was 8.38% ± 0.56%. The surface morphology of NCTD nanoparticles (NPs) coated with PVP K(30) was characterized using various analytical techniques, including X-ray diffraction and atomic force microscopy. NCTD and its metabolites were analyzed using a sensitive and specific liquid chromatography-tandem mass spectrometry method with samples from mice and rats. The results indicated the importance of the PVP coating in controlling the shape and improving the entrapment efficiency of the NPs. Pharmacokinetic profiles of the NCTD group and PVP-NCTD-NP group, after oral and intravenous administration in rats, revealed that relative bioavailabilities were 173.3% and 325.5%, respectively. The elimination half-life increased, and there was an obvious decrease in clearance. The tissue distribution of NCTD in mice after the intravenous administration of both formulations was investigated. The drug was not quantifiable at 6 hours in all tissues except for the liver and kidneys. The distribution of the drug in the liver and bile was notably improved in the PVP-NCTD-NP group. The metabolites and excretion properties of NCTD were investigated by analyzing rat feces and urine samples, collected after oral administration. A prototype drug and two metabolites were found in the feces, and seven metabolites in the urine. The primary elimination route of NCTD was via the urine. The quantity of the parent drug eliminated in the feces of the PVP-NCTD-NP group, was 32 times greater than that of the NCTD group, indicating that the NPs dramatically increased the reduction quantity from liver to bile. We conclude that PVP-NCTD-NPs are an adequate formulation for enhancing the absorption of NCTD, and significantly improving therapeutic effects targeting the hepatic system. Decarboxylation and hydroxylation were the dominant metabolic pathways for NCTD. Metabolites were mainly excreted into rat kidney and finally into urine.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Nanoparticles/chemistry , Animals , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Chitosan/chemistry , Chromatography, Liquid , Female , Male , Mice , Mice, Inbred ICR , Microscopy, Atomic Force , Nanomedicine , Nanoparticles/ultrastructure , Povidone/chemistry , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry , Tissue Distribution
7.
Int J Nanomedicine ; 7: 1819-27, 2012.
Article in English | MEDLINE | ID: mdl-22619530

ABSTRACT

In this paper, two novel liver-targeting nanoparticles, norcantharidin-loaded chitosan nanoparticles (NCTD-CS-NPs) and norcantharidin-associated galactosylated chitosan nanoparticles (NCTD-GC-NPs), were prepared using ionic cross-linkage. The physical properties, particle size, encapsulation efficiency, and drug release characteristics of the nanoparticles were investigated in vitro. To investigate the intestinal absorption mechanisms of the two preparations, a series of experiments was carried out, including in situ circulation method, in vitro everted gut sacs, and Ussing chamber perfusion technique. The absorption rate constants (Ka) of NCTD at different segments were found to be duodenum > jejunum > ileum > colon. The concentration had no distinctive effect on absorption kinetics, suggesting that drug absorption is not dose-dependent. The transport of NCTD was found to be inhibited by P-glycoprotein (P-gp) inhibitor, indicating that NCTD might be the substrate of P-gp. The order of the absorption enhancer effects were as follows: low molecular weight chitosan (CS-8kDa) > high molecular weight chitosan (CS-30kDa) > Poloxamer > sodium dodecyl sulfate (SDS) > sodium deoxycholate (SDCh). The results indicate that the chitosan nanoparticles can improve intestinal absorption of NCTD.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Intestinal Absorption , Nanoparticles/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Chitosan/chemistry , Drug Carriers/chemistry , Drug Delivery Systems , In Vitro Techniques , Liver/drug effects , Liver/metabolism , Male , Microscopy, Electron, Transmission , Nanomedicine , Nanoparticles/ultrastructure , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...