Cargando…

Co-administration of temozolomide (TMZ) and the experimental therapeutic targeting miR-10b, profoundly affects the tumorigenic phenotype of human glioblastoma cells

Introduction: Recent studies have shown that miRNA-10b is highly expressed in high-grade glioblastoma multiforme (GBM), and its inhibition leads to deregulation of multiple pathways in tumorigenesis, resulting in repression of tumor growth and increased apoptosis. Thus, we hypothesized that suppress...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Ming, Kim, Bryan, Robertson, Neil, Mondal, Sujan Kumar, Medarova, Zdravka, Moore, Anna
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10311069/
https://www.ncbi.nlm.nih.gov/pubmed/37398551
http://dx.doi.org/10.3389/fmolb.2023.1179343
_version_ 1785066667057872896
author Chen, Ming
Kim, Bryan
Robertson, Neil
Mondal, Sujan Kumar
Medarova, Zdravka
Moore, Anna
author_facet Chen, Ming
Kim, Bryan
Robertson, Neil
Mondal, Sujan Kumar
Medarova, Zdravka
Moore, Anna
author_sort Chen, Ming
collection PubMed
description Introduction: Recent studies have shown that miRNA-10b is highly expressed in high-grade glioblastoma multiforme (GBM), and its inhibition leads to deregulation of multiple pathways in tumorigenesis, resulting in repression of tumor growth and increased apoptosis. Thus, we hypothesized that suppressing miR-10b could enhance the cytotoxicity of conventional GBM chemotherapy with temozolomide (TMZ). Methods: Inhibition of miR-10b in glioblastoma cells was achieved using an experimental therapeutic consisting of anti-miR10b antagomirs conjugated to iron oxide nanoparticles (termed MN-anti-miR10b). The nanoparticles serve as delivery vehicles for the antagomirs as well as imaging reporters guiding the delivery in future animal studies. Results: Treatment of U251 and LN229 human glioblastoma cells with MN-anti-miR10b led to inhibition of miR-10b accompanied by repression of growth and increase in apoptosis. We next explored whether MN-anti-miR10b could enhance the cytotoxic effect of TMZ. During these studies, we unexpectedly found that TMZ monotherapy increased miR-10b expression and changed the expression of corresponding miR-10b targets. This discovery led to the design of a sequence-dependent combination treatment, in which miR-10b inhibition and induction of apoptosis by MN-anti-miR10b was followed by a sub-therapeutic dose of TMZ, which caused cell cycle arrest and ultimately cell death. This combination was highly successful in significant enhancement of apoptosis and decrease in cell migration and invasiveness. Discussion: Considering the unexpected effects of TMZ on miR-10b expression and possible implications on its clinical application, we reasoned that comprehensive in vitro studies were warranted before embarking on studies in animals. These intriguing findings serve as a solid foundation for future in vivo studies and offer promise for the successful treatment of GBM.
format Online
Article
Text
id pubmed-10311069
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-103110692023-07-01 Co-administration of temozolomide (TMZ) and the experimental therapeutic targeting miR-10b, profoundly affects the tumorigenic phenotype of human glioblastoma cells Chen, Ming Kim, Bryan Robertson, Neil Mondal, Sujan Kumar Medarova, Zdravka Moore, Anna Front Mol Biosci Molecular Biosciences Introduction: Recent studies have shown that miRNA-10b is highly expressed in high-grade glioblastoma multiforme (GBM), and its inhibition leads to deregulation of multiple pathways in tumorigenesis, resulting in repression of tumor growth and increased apoptosis. Thus, we hypothesized that suppressing miR-10b could enhance the cytotoxicity of conventional GBM chemotherapy with temozolomide (TMZ). Methods: Inhibition of miR-10b in glioblastoma cells was achieved using an experimental therapeutic consisting of anti-miR10b antagomirs conjugated to iron oxide nanoparticles (termed MN-anti-miR10b). The nanoparticles serve as delivery vehicles for the antagomirs as well as imaging reporters guiding the delivery in future animal studies. Results: Treatment of U251 and LN229 human glioblastoma cells with MN-anti-miR10b led to inhibition of miR-10b accompanied by repression of growth and increase in apoptosis. We next explored whether MN-anti-miR10b could enhance the cytotoxic effect of TMZ. During these studies, we unexpectedly found that TMZ monotherapy increased miR-10b expression and changed the expression of corresponding miR-10b targets. This discovery led to the design of a sequence-dependent combination treatment, in which miR-10b inhibition and induction of apoptosis by MN-anti-miR10b was followed by a sub-therapeutic dose of TMZ, which caused cell cycle arrest and ultimately cell death. This combination was highly successful in significant enhancement of apoptosis and decrease in cell migration and invasiveness. Discussion: Considering the unexpected effects of TMZ on miR-10b expression and possible implications on its clinical application, we reasoned that comprehensive in vitro studies were warranted before embarking on studies in animals. These intriguing findings serve as a solid foundation for future in vivo studies and offer promise for the successful treatment of GBM. Frontiers Media S.A. 2023-06-15 /pmc/articles/PMC10311069/ /pubmed/37398551 http://dx.doi.org/10.3389/fmolb.2023.1179343 Text en Copyright © 2023 Chen, Kim, Robertson, Mondal, Medarova and Moore. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Molecular Biosciences
Chen, Ming
Kim, Bryan
Robertson, Neil
Mondal, Sujan Kumar
Medarova, Zdravka
Moore, Anna
Co-administration of temozolomide (TMZ) and the experimental therapeutic targeting miR-10b, profoundly affects the tumorigenic phenotype of human glioblastoma cells
title Co-administration of temozolomide (TMZ) and the experimental therapeutic targeting miR-10b, profoundly affects the tumorigenic phenotype of human glioblastoma cells
title_full Co-administration of temozolomide (TMZ) and the experimental therapeutic targeting miR-10b, profoundly affects the tumorigenic phenotype of human glioblastoma cells
title_fullStr Co-administration of temozolomide (TMZ) and the experimental therapeutic targeting miR-10b, profoundly affects the tumorigenic phenotype of human glioblastoma cells
title_full_unstemmed Co-administration of temozolomide (TMZ) and the experimental therapeutic targeting miR-10b, profoundly affects the tumorigenic phenotype of human glioblastoma cells
title_short Co-administration of temozolomide (TMZ) and the experimental therapeutic targeting miR-10b, profoundly affects the tumorigenic phenotype of human glioblastoma cells
title_sort co-administration of temozolomide (tmz) and the experimental therapeutic targeting mir-10b, profoundly affects the tumorigenic phenotype of human glioblastoma cells
topic Molecular Biosciences
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10311069/
https://www.ncbi.nlm.nih.gov/pubmed/37398551
http://dx.doi.org/10.3389/fmolb.2023.1179343
work_keys_str_mv AT chenming coadministrationoftemozolomidetmzandtheexperimentaltherapeutictargetingmir10bprofoundlyaffectsthetumorigenicphenotypeofhumanglioblastomacells
AT kimbryan coadministrationoftemozolomidetmzandtheexperimentaltherapeutictargetingmir10bprofoundlyaffectsthetumorigenicphenotypeofhumanglioblastomacells
AT robertsonneil coadministrationoftemozolomidetmzandtheexperimentaltherapeutictargetingmir10bprofoundlyaffectsthetumorigenicphenotypeofhumanglioblastomacells
AT mondalsujankumar coadministrationoftemozolomidetmzandtheexperimentaltherapeutictargetingmir10bprofoundlyaffectsthetumorigenicphenotypeofhumanglioblastomacells
AT medarovazdravka coadministrationoftemozolomidetmzandtheexperimentaltherapeutictargetingmir10bprofoundlyaffectsthetumorigenicphenotypeofhumanglioblastomacells
AT mooreanna coadministrationoftemozolomidetmzandtheexperimentaltherapeutictargetingmir10bprofoundlyaffectsthetumorigenicphenotypeofhumanglioblastomacells