HDAC1 and HDAC6 are essential for driving growth in IDH1 mutant glioma

Abstract Low-grade and secondary high-grade gliomas frequently contain mutations in the IDH1 or IDH2 metabolic enzymes that are hypothesized to drive tumorigenesis by inhibiting many of the chromatin-regulating enzymes that regulate DNA structure. Histone deacetylase inhibitors are promising anti-ca...

Full description

Bibliographic Details
Main Authors: Matthew C. Garrett, Rebecca Albano, Troy Carnwath, Lubayna Elahi, Catherine A. Behrmann, Merissa Pemberton, Daniel Woo, Eric O’Brien, Brett VanCauwenbergh, John Perentesis, Sanjit Shah, Matthew Hagan, Ady Kendler, Chuntao Zhao, Aditi Paranjpe, Krishna Roskin, Harley Kornblum, David R. Plas, Q. Richard Lu
Format: Article
Language:English
Published: Nature Portfolio 2023-08-01
Series:Scientific Reports
Online Access:https://doi.org/10.1038/s41598-023-33889-3
_version_ 1797752796505702400
author Matthew C. Garrett
Rebecca Albano
Troy Carnwath
Lubayna Elahi
Catherine A. Behrmann
Merissa Pemberton
Daniel Woo
Eric O’Brien
Brett VanCauwenbergh
John Perentesis
Sanjit Shah
Matthew Hagan
Ady Kendler
Chuntao Zhao
Aditi Paranjpe
Krishna Roskin
Harley Kornblum
David R. Plas
Q. Richard Lu
author_facet Matthew C. Garrett
Rebecca Albano
Troy Carnwath
Lubayna Elahi
Catherine A. Behrmann
Merissa Pemberton
Daniel Woo
Eric O’Brien
Brett VanCauwenbergh
John Perentesis
Sanjit Shah
Matthew Hagan
Ady Kendler
Chuntao Zhao
Aditi Paranjpe
Krishna Roskin
Harley Kornblum
David R. Plas
Q. Richard Lu
author_sort Matthew C. Garrett
collection DOAJ
description Abstract Low-grade and secondary high-grade gliomas frequently contain mutations in the IDH1 or IDH2 metabolic enzymes that are hypothesized to drive tumorigenesis by inhibiting many of the chromatin-regulating enzymes that regulate DNA structure. Histone deacetylase inhibitors are promising anti-cancer agents and have already been used in clinical trials. However, a clear understanding of their mechanism or gene targets is lacking. In this study, the authors genetically dissect patient-derived IDH1 mutant cultures to determine which HDAC enzymes drive growth in IDH1 mutant gliomas. A panel of patient-derived gliomasphere cell lines (2 IDH1 mutant lines, 3 IDH1 wildtype lines) were subjected to a drug-screen of epigenetic modifying drugs from different epigenetic classes. The effect of LBH (panobinostat) on gene expression and chromatin structure was tested on patient-derived IDH1 mutant lines. The role of each of the highly expressed HDAC enzymes was molecularly dissected using lentiviral RNA interference knock-down vectors and a patient-derived IDH1 mutant in vitro model of glioblastoma (HK252). These results were then confirmed in an in vivo xenotransplant model (BT-142). The IDH1 mutation leads to gene down-regulation, DNA hypermethylation, increased DNA accessibility and H3K27 hypo-acetylation in two distinct IDH1 mutant over-expression models. The drug screen identified histone deacetylase inhibitors (HDACi) and panobinostat (LBH) more specifically as the most selective compounds to inhibit growth in IDH1 mutant glioma lines. Of the eleven annotated HDAC enzymes (HDAC1-11) only six are expressed in IDH1 mutant glioma tissue samples and patient-derived gliomasphere lines (HDAC1-4, HDAC6, and HDAC9). Lentiviral knock-down experiments revealed that HDAC1 and HDAC6 are the most consistently essential for growth both in vitro and in vivo and target very different gene modules. Knock-down of HDAC1 or HDAC6 in vivo led to a more circumscribed less invasive tumor. The gene dysregulation induced by the IDH1 mutation is wide-spread and only partially reversible by direct IDH1 inhibition. This study identifies HDAC1 and HDAC6 as important and drug-targetable enzymes that are necessary for growth and invasiveness in IDH1 mutant gliomas.
first_indexed 2024-03-12T17:09:39Z
format Article
id doaj.art-cd41171c4c2c402c887577588bb94c39
institution Directory Open Access Journal
issn 2045-2322
language English
last_indexed 2024-03-12T17:09:39Z
publishDate 2023-08-01
publisher Nature Portfolio
record_format Article
series Scientific Reports
spelling doaj.art-cd41171c4c2c402c887577588bb94c392023-08-06T11:11:16ZengNature PortfolioScientific Reports2045-23222023-08-0113111210.1038/s41598-023-33889-3HDAC1 and HDAC6 are essential for driving growth in IDH1 mutant gliomaMatthew C. Garrett0Rebecca Albano1Troy Carnwath2Lubayna Elahi3Catherine A. Behrmann4Merissa Pemberton5Daniel Woo6Eric O’Brien7Brett VanCauwenbergh8John Perentesis9Sanjit Shah10Matthew Hagan11Ady Kendler12Chuntao Zhao13Aditi Paranjpe14Krishna Roskin15Harley Kornblum16David R. Plas17Q. Richard Lu18Department of Neurosurgery, University of Cincinnati College of MedicineDepartment of Neurosurgery, University of Cincinnati College of MedicineUniversity of Cincinnati College of MedicineDepartment of Molecular Cell and Developmental Biology, University of California Los AngelesDepartment of Cancer Biology, University of CincinnatiUniversity of Cincinnati College of MedicineDepartment of Neurology, University of Cincinnati College of MedicineDivision of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of MedicineDivision of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of MedicineDivision of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of MedicineDepartment of Neurosurgery, University of Cincinnati College of MedicineDepartment of Pathology and Laboratory Medicine, University of Cincinnati College of MedicineDepartment of Pathology and Laboratory Medicine, University of Cincinnati College of MedicineDivision of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of MedicineBioinformatics Collaborative Services, Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical CenterBioinformatics Collaborative Services, Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical CenterDepartment of Molecular Cell and Developmental Biology, University of California Los AngelesDepartment of Cancer Biology, University of CincinnatiDivision of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of MedicineAbstract Low-grade and secondary high-grade gliomas frequently contain mutations in the IDH1 or IDH2 metabolic enzymes that are hypothesized to drive tumorigenesis by inhibiting many of the chromatin-regulating enzymes that regulate DNA structure. Histone deacetylase inhibitors are promising anti-cancer agents and have already been used in clinical trials. However, a clear understanding of their mechanism or gene targets is lacking. In this study, the authors genetically dissect patient-derived IDH1 mutant cultures to determine which HDAC enzymes drive growth in IDH1 mutant gliomas. A panel of patient-derived gliomasphere cell lines (2 IDH1 mutant lines, 3 IDH1 wildtype lines) were subjected to a drug-screen of epigenetic modifying drugs from different epigenetic classes. The effect of LBH (panobinostat) on gene expression and chromatin structure was tested on patient-derived IDH1 mutant lines. The role of each of the highly expressed HDAC enzymes was molecularly dissected using lentiviral RNA interference knock-down vectors and a patient-derived IDH1 mutant in vitro model of glioblastoma (HK252). These results were then confirmed in an in vivo xenotransplant model (BT-142). The IDH1 mutation leads to gene down-regulation, DNA hypermethylation, increased DNA accessibility and H3K27 hypo-acetylation in two distinct IDH1 mutant over-expression models. The drug screen identified histone deacetylase inhibitors (HDACi) and panobinostat (LBH) more specifically as the most selective compounds to inhibit growth in IDH1 mutant glioma lines. Of the eleven annotated HDAC enzymes (HDAC1-11) only six are expressed in IDH1 mutant glioma tissue samples and patient-derived gliomasphere lines (HDAC1-4, HDAC6, and HDAC9). Lentiviral knock-down experiments revealed that HDAC1 and HDAC6 are the most consistently essential for growth both in vitro and in vivo and target very different gene modules. Knock-down of HDAC1 or HDAC6 in vivo led to a more circumscribed less invasive tumor. The gene dysregulation induced by the IDH1 mutation is wide-spread and only partially reversible by direct IDH1 inhibition. This study identifies HDAC1 and HDAC6 as important and drug-targetable enzymes that are necessary for growth and invasiveness in IDH1 mutant gliomas.https://doi.org/10.1038/s41598-023-33889-3
spellingShingle Matthew C. Garrett
Rebecca Albano
Troy Carnwath
Lubayna Elahi
Catherine A. Behrmann
Merissa Pemberton
Daniel Woo
Eric O’Brien
Brett VanCauwenbergh
John Perentesis
Sanjit Shah
Matthew Hagan
Ady Kendler
Chuntao Zhao
Aditi Paranjpe
Krishna Roskin
Harley Kornblum
David R. Plas
Q. Richard Lu
HDAC1 and HDAC6 are essential for driving growth in IDH1 mutant glioma
Scientific Reports
title HDAC1 and HDAC6 are essential for driving growth in IDH1 mutant glioma
title_full HDAC1 and HDAC6 are essential for driving growth in IDH1 mutant glioma
title_fullStr HDAC1 and HDAC6 are essential for driving growth in IDH1 mutant glioma
title_full_unstemmed HDAC1 and HDAC6 are essential for driving growth in IDH1 mutant glioma
title_short HDAC1 and HDAC6 are essential for driving growth in IDH1 mutant glioma
title_sort hdac1 and hdac6 are essential for driving growth in idh1 mutant glioma
url https://doi.org/10.1038/s41598-023-33889-3
work_keys_str_mv AT matthewcgarrett hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT rebeccaalbano hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT troycarnwath hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT lubaynaelahi hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT catherineabehrmann hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT merissapemberton hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT danielwoo hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT ericobrien hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT brettvancauwenbergh hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT johnperentesis hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT sanjitshah hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT matthewhagan hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT adykendler hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT chuntaozhao hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT aditiparanjpe hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT krishnaroskin hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT harleykornblum hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT davidrplas hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma
AT qrichardlu hdac1andhdac6areessentialfordrivinggrowthinidh1mutantglioma