Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters











Database
Language
Publication year range
1.
Int J Mol Sci ; 21(4)2020 Feb 22.
Article in English | MEDLINE | ID: mdl-32098295

ABSTRACT

Glioblastoma multiforme (GBM) is an aggressive adult brain tumour with poor prognosis. Roles for peptidylarginine deiminases (PADs) in GBM have recently been highlighted. Here, two GBM cell lines were treated with PAD2, PAD3 and PAD4 isozyme-specific inhibitors. Effects were assessed on extracellular vesicle (EV) signatures, including EV-microRNA cargo (miR21, miR126 and miR210), and on changes in cellular protein expression relevant for mitochondrial housekeeping (prohibitin (PHB)) and cancer progression (stromal interaction molecule 1 (STIM-1) and moesin), as well as assessing cell invasion. Overall, GBM cell-line specific differences for the three PAD isozyme-specific inhibitors were observed on modulation of EV-signatures, PHB, STIM-1 and moesin protein levels, as well as on cell invasion. The PAD3 inhibitor was most effective in modulating EVs to anti-oncogenic signatures (reduced miR21 and miR210, and elevated miR126), to reduce cell invasion and to modulate protein expression of pro-GBM proteins in LN229 cells, while the PAD2 and PAD4 inhibitors were more effective in LN18 cells. Furthermore, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways for deiminated proteins relating to cancer, metabolism and inflammation differed between the two GBM cell lines. Our findings highlight roles for the different PAD isozymes in the heterogeneity of GBM tumours and the potential for tailored PAD-isozyme specific treatment.


Subject(s)
Enzyme Inhibitors/pharmacology , Extracellular Vesicles/genetics , Gene Expression Regulation, Neoplastic/drug effects , MicroRNAs/genetics , Protein-Arginine Deiminase Type 2/antagonists & inhibitors , Protein-Arginine Deiminase Type 3/antagonists & inhibitors , Protein-Arginine Deiminase Type 4/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Extracellular Vesicles/metabolism , Humans , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Prohibitins , Protein-Arginine Deiminase Type 2/metabolism , Protein-Arginine Deiminase Type 3/metabolism , Protein-Arginine Deiminase Type 4/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism
2.
Transl Oncol ; 12(3): 513-522, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30597288

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and aggressive form of primary malignant brain tumor in adults, with poor prognosis. Extracellular vesicles (EVs) are key-mediators for cellular communication through transfer of proteins and genetic material. Cancers, such as GBM, use EV release for drug-efflux, pro-oncogenic signaling, invasion and immunosuppression; thus the modulation of EV release and cargo is of considerable clinical relevance. As EV-inhibitors have been shown to increase sensitivity of cancer cells to chemotherapy, and we recently showed that cannabidiol (CBD) is such an EV-modulator, we investigated whether CBD affects EV profile in GBM cells in the presence and absence of temozolomide (TMZ). Compared to controls, CBD-treated cells released EVs containing lower levels of pro-oncogenic miR21 and increased levels of anti-oncogenic miR126; these effects were greater than with TMZ alone. In addition, prohibitin (PHB), a multifunctional protein with mitochondrial protective properties and chemoresistant functions, was reduced in GBM cells following 1 h CBD treatment. This data suggests that CBD may, via modulation of EVs and PHB, act as an adjunct to enhance treatment efficacy in GBM, supporting evidence for efficacy of cannabinoids in GBM.

3.
Int J Mol Sci ; 20(1)2018 Dec 28.
Article in English | MEDLINE | ID: mdl-30597867

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive form of adult primary malignant brain tumour with poor prognosis. Extracellular vesicles (EVs) are a key-mediator through which GBM cells promote a pro-oncogenic microenvironment. Peptidylarginine deiminases (PADs), which catalyze the post-translational protein deimination of target proteins, are implicated in cancer, including via EV modulation. Pan-PAD inhibitor Cl-amidine affected EV release from GBM cells, and EV related microRNA cargo, with reduced pro-oncogenic microRNA21 and increased anti-oncogenic microRNA126, also in combinatory treatment with the chemotherapeutic agent temozolomide (TMZ). The GBM cell lines under study, LN18 and LN229, differed in PAD2, PAD3 and PAD4 isozyme expression. Various cytoskeletal, nuclear and mitochondrial proteins were identified to be deiminated in GBM, including prohibitin (PHB), a key protein in mitochondrial integrity and also involved in chemo-resistance. Post-translational deimination of PHB, and PHB protein levels, were reduced after 1 h treatment with pan-PAD inhibitor Cl-amidine in GBM cells. Histone H3 deimination was also reduced following Cl-amidine treatment. Multifaceted roles for PADs on EV-mediated pathways, as well as deimination of mitochondrial, nuclear and invadopodia related proteins, highlight PADs as novel targets for modulating GBM tumour communication.


Subject(s)
Extracellular Vesicles/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , MicroRNAs/metabolism , Protein-Arginine Deiminases/metabolism , Antineoplastic Agents/pharmacology , Biological Transport , Cell Line, Tumor , Cell Survival , Chromatography, Liquid , Extracellular Vesicles/ultrastructure , Histones/metabolism , Humans , MicroRNAs/genetics , Ornithine/analogs & derivatives , Ornithine/pharmacology , Prohibitins , Protein Processing, Post-Translational/drug effects , Protein-Arginine Deiminases/genetics , Proteome , Proteomics/methods , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL