scholarly journals DIPG-58. HISTONE H3 WILD-TYPE DIPG/DMG OVEREXPRESSING EZHIP EXTEND THE SPECTRUM OF DIFFUSE MIDLINE GLIOMAS WITH PRC2 INHIBITION BEYOND H3-K27M MUTATION

2020 ◽  
Vol 22 (Supplement_3) ◽  
pp. iii298-iii298
Author(s):  
David Castel ◽  
Thomas Kergrohen ◽  
Arnault Tauziède-Espariat ◽  
Alan Mackay ◽  
Samia Ghermaoui ◽  
...  

Abstract Diffuse midline gliomas (DMG) H3 K27M-mutant were introduced in the 2016 WHO Classification unifying diffuse intrinsic pontine gliomas (DIPG) and gliomas from the thalamus and spinal cord harboring a histone H3-K27M mutation leading to Polycomb Repressor Complex 2 (PRC2) inhibition. However, few cases of DMG tumors presenting a H3K27 trimethylation loss, but lacking an H3-K27M mutation were reported. To address this question, we combined a retrospective cohort of 10 patients biopsied for a DIPG at the Necker Hospital or included in the BIOMEDE trial (NCT02233049) and extended our analysis to H3-wildtype (WT) diffuse gliomas from other midline locations presenting either H3K27 trimethylation loss or ACVR1 mutation from Necker, ICR, the HERBY trial, the INFORM registry study and the St. Jude PCGP representing 9 additional cases. Genomic profiling identified alterations frequently found in DMG, but none could explain the observed loss of H3K27 trimethylation. Similar observations were previously made in the PF-A subgroup of ependymoma, where the H3K27me3 loss resulted from EZHIP/CXorf67 overexpression rather than H3-K27M mutations. We thus analyzed EZHIP expression and observed its overexpression in all but one H3-WT DMGs compared to H3-K27M mutated tumors (EZHIP negative). Strikingly, based on their DNA methylation profiles, all H3-WT DMG samples analyzed clustered close to H3-K27M DIPG, rather than EZHIP overexpressing PF-A ependymomas. To conclude, we described a new subgroup of DMG lacking H3-K27M mutation, defined by H3K27 trimethylation loss and EZHIP overexpression that can be detected by IHC. We propose that these EZHIP/H3-WT DMGs extend the spectrum of DMG with PRC2 inhibition beyond H3-K27M mutation.

2020 ◽  
Vol 79 (10) ◽  
pp. 1038-1043
Author(s):  
Meaghan Morris ◽  
Meghan Driscoll ◽  
John W Henson ◽  
Charles Cobbs ◽  
LiQun Jiang ◽  
...  

Abstract Mutations in histone H3 are key molecular drivers of pediatric and young adult high-grade gliomas. Histone H3 G34R mutations occur in hemispheric high-grade gliomas and H3 K27M mutations occur in aggressive, though histologically diverse, midline gliomas. Here, we report 2 rare cases of histologically low-grade gliomas with gemistocytic morphology and sequencing-confirmed histone H3 G34R mutations. One case is a histologically low-grade gemistocytic astrocytoma with a G34R-mutation in H3F3A. The second case is a histologically low-grade gemistocytic astrocytoma with co-occurring K27M and G34R mutations in HIST1H3B. Review of prior histone H3-mutant gliomas sequenced at our institution shows a divergent clinical and immunohistochemical pattern in the 2 cases. The first case is similar to prior histone H3 G34R-mutant tumors, while the second case most closely resembles prior histone H3 K27M-mutant gliomas. These represent novel cases of sequencing-confirmed histone H3 G34R-mutant gliomas with low-grade histology and add to the known rare cases of G34R-mutant tumors with gemistocytic morphology. Although K27M and G34R mutations are thought to be mutually exclusive, we document combined K27M and G34R mutations in HIST1H3B and present evidence suggesting the K27M-mutation drove tumor phenotype in this dual mutant glioma.


CNS Oncology ◽  
2021 ◽  
pp. CNS71
Author(s):  
Justin Thomas Low ◽  
Shih-Hsiu Wang ◽  
Katherine B Peters

Diffuse midline gliomas harboring histone H3 K27M mutations are most commonly found in the brainstem of children. This mutation confers a WHO grade IV designation and is associated with a particularly poor prognosis. Although traditionally considered to be a disease of children and young adults, a number of recent reports have described H3 K27M mutations in older adults with diffuse midline gliomas. Here, we present the unusual case of a diffuse midline glioma in the pons and cerebellum of an 83-year-old woman and review the evolving clinical literature on this entity in adults. This case underscores that it may occur even in older adults, in whom prognostic and treatment paradigms used in pediatrics may not be directly applicable.


2020 ◽  
Vol 139 (6) ◽  
pp. 1109-1113 ◽  
Author(s):  
David Castel ◽  
Thomas Kergrohen ◽  
Arnault Tauziède-Espariat ◽  
Alan Mackay ◽  
Samia Ghermaoui ◽  
...  
Keyword(s):  

2021 ◽  
Vol 7 (4) ◽  
pp. eabe2299 ◽  
Author(s):  
Na Wang ◽  
Jonathan I. Gent ◽  
R. Kelly Dawe

The production of haploids is an important first step in creating many new plant varieties. One approach used in Arabidopsis involves crossing plants expressing different forms of centromeric histone H3 (CENP-A/CENH3) and subsequent loss of genome with weaker centromeres. However, the method has been ineffective in crop plants. Here, we describe a greatly simplified method based on crossing maize lines that are heterozygous for a cenh3 null mutation. Crossing +/cenh3 to wild-type plants in both directions yielded haploid progeny. Genome elimination was determined by the cenh3 genotype of the gametophyte, suggesting that centromere failure is caused by CENH3 dilution during the postmeiotic cell divisions that precede gamete formation. The cenh3 haploid inducer works as a vigorous hybrid and can be transferred to other lines in a single cross, making it versatile for a variety of applications.


1997 ◽  
Vol 17 (11) ◽  
pp. 6303-6310 ◽  
Author(s):  
L Yu ◽  
M A Gorovsky

Although quantitatively minor replication-independent (replacement) histone variants have been found in a wide variety of organisms, their functions remain unknown. Like the H3.3 replacement variants of vertebrates, hv2, an H3 variant in the ciliated protozoan Tetrahymena thermophila, is synthesized and deposited in nuclei of nongrowing cells. Although hv2 is clearly an H3.3-like replacement variant by its expression, sequence analysis indicates that it evolved independently of the H3.3 variants of multicellular eukaryotes. This suggested that it is the constitutive synthesis, not the particular protein sequence, of these variants that is important in the function of H3 replacement variants. Here, we demonstrate that the gene (HHT3) encoding hv2 or either gene (HHT1 or HHT2) encoding the abundant major H3 can be completely knocked out in Tetrahymena. Surprisingly, when cells lacking hv2 are starved, a major histone H3 mRNA transcribed by the HHT2 gene, which is synthesized little, if at all, in wild-type nongrowing cells, is easily detectable. Both HHT2 and HHT3 knockout strains show no obvious defect during vegetative growth. In addition, a mutant with the double knockout of HHT1 and HHT3 is viable while the HHT2 HHT3 double-knockout mutant is not. These results argue strongly that cells require a constitutively expressed H3 gene but that the particular sequence being expressed is not critical.


Genetics ◽  
1991 ◽  
Vol 128 (1) ◽  
pp. 29-35
Author(s):  
D N Arvidson ◽  
M Shapiro ◽  
P Youderian

Abstract The Escherichia coli trpR gene encodes tryptophan aporepressor, which binds the corepressor ligand, L-tryptophan, to form an active repressor complex. The side chain of residue valine 58 of Trp aporepressor sits at the bottom of the corepressor (L-tryptophan) binding pocket. Mutant trpR genes encoding changes of Val58 to the other 19 naturally occurring amino acids were made. Each of the mutant proteins requires a higher intracellular concentration of tryptophan for activation of DNA binding than wild-type aporepressor. Whereas wild-type aporepressor is activated better by 5-methyltryptophan (5-MT) than by tryptophan, Ile58 and other mutant aporepressors prefer tryptophan to 5-MT as corepressor, and Ala58 and Gly58 prefer 5-MT much more strongly than wild-type aporepressor in vivo. These mutant aporepressors are the first examples of DNA-binding proteins with altered specificities of cofactor recognition.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3759-3759
Author(s):  
Jaesung Seo ◽  
Li Li ◽  
Donald Small

Mutations of DNMT3A are one the most frequently observed alterations in AML patients. The DNMT3A R882 mutation appears to confer a dominant-negative loss-of function effect and changes the DNA binding preference according to recent studies. DNMT3A R882 mutations are also found at increasing frequency with age in healthy elderly populations and are one of the earliest pre-malignant alterations in the clonal evolution progression to leukemia. Studies have shown that DNMT3A mutations decrease overall DNA methylation and through less clear mechanisms, also impact the epigenetic landscape by effecting changes in histone acetylation. Here we investigated potential mechanisms by which loss of DNMT3A activity changes histone acetylation. DNMT3A binds with many proteins that regulate chromatin biology and gene transcription. Among those interacting proteins, we focused on the DNMT3A-HDAC interaction and their regulation of target gene suppression. To investigate whether there are differences in binding of wild-type vs. mutant DNMT3A to HDACs, we performed immunoprecipitation and Western blotting assays using Myc- tagged wild-type and R882 mutated DNMT3A. We found that DNMT3A R882 mutants showed reduced interaction with HDAC1 and 2. In addition, upon treatment with HDAC inhibitors (HDACi), DNMT3A mutant protein was more easily dissociated from HDAC1/2 than was wild-type DNMT3A. Intriguingly, covalent modification of DNMT3A R882 by SUMO1 protein was significantly enhanced relative to wild type DNMT3A. Together, we suggest that the weak complex formation between mutant DNMT3A and HDACs results from augmented SUMOylation of the R882 mutant. Because the DNMT3A R882 mutation reduces its methyl transferase activity, we investigated which genes would be upregulated from the DNMT3A repressor complex. To do this, we established isogenic TF-1 cell lines that harbor haploid DNMT3A knockout (DNMT3A+/-) using the Cripsr-Cas9 system. We also treated cells with HDACi and 5-azacytidine (5-aza) which inhibit HDAC and DNMT, respectively. Interestingly, we discovered that PD-L1 expression is induced by HDACi and 5-aza treatment. Chemical inhibition by 5-aza or genetic inhibition by knockout reduces DNMT3A activity and synergized with HDACi to increase PD-L1 expression. Flow cytometry analysis also demonstrated increased membrane PD-L1 expression in response to HDACi. We also found out that DNMT3A+/- resulted in higher Histone H3K27 acetylation, which is known as a gene activation mark. Higher H3K27 acetylation in DNMT3A+/- cell confirms the findings by other groups but the mechanisms by which this occurs are unknown. We suggest that haploinsufficiency of DNMT3A results in a reduced DNMT3A-HDAC interaction leading to higher H3K27 acetylation and increased PD-L1 expression. Our results also revealed that HDACi treatment induced cell cycle arrest, DNA damage and apoptosis at increasing levels in DNMT3A+/- cell. Even though the DNMT3A+/- TF-1 showed increased sensitivity to HDACi treatment, we observed a correlation of higher phosphor- ERK1/2 and PD-L1 levels in the surviving cells. The enhanced expression of PD-L1 and activation of ERK1/2 may explain in part how mutated DNMT3A contributes to drug resistance and immune checkpoint avoidance. Many oncology clinical trials are underway utilizing HDACi. However, the questions of which mutational backgrounds might be most sensitive to these agents and how to best combine them with other agents remain to be answered. To test whether reduced DNMT3A activity increases PD-L1 expression in vivo, we crossed floxed DNMT3A mice with Mx1-Cre mice. After 4 weeks of induction of Cre recombinase by injecting pIpC in the progeny carrying both genetically engineered changes, lineage depleted mouse BM cells were analyzed for PD-L1 expression using quantitative RT-PCR. BM cells derived from DNMT3A knockout mice showed increased expression of PD-L1 compared to wild-type mice. Treatment of these BM cells with an HDACi and/or 5-aza resulted in a synergistic induction of PD-L1 expression for the combination. Taken together, we suggest that mutant DNMT3A induces higher H3K27 acetylation along with PD-L1 expression due to a looser complex between HDAC1 and mutant DNMT3A. Therefore, we suggest that combined treatment with an HDACi and an immune checkpoint inhibitor targeting the PD-L1/PD-1 axis might be a promising strategy for treating DNMT3A mutant AML patients. Disclosures Small: InSilico Medicine: Membership on an entity's Board of Directors or advisory committees; Pharos I, B & T: Consultancy, Research Funding.


2019 ◽  
Author(s):  
Teresa W. Lee ◽  
Heidi S. David ◽  
Amanda K. Engstrom ◽  
Brandon S. Carpenter ◽  
David J. Katz

ABSTRACTDuring active transcription, the COMPASS complex methylates histone H3 at lysine 4 (H3K4me). In Caenorhabditis elegans, mutations in COMPASS subunits, including WDR-5, extend lifespan and enable the inheritance of increased lifespan in wild-type descendants. Here we show that the increased lifespan of wdr-5 mutants is itself a transgenerational trait that manifests after eighteen generations and correlates with changes in the heterochromatin factor H3K9me2. Additionally, we find that wdr-5 mutant longevity and its inheritance requires the H3K9me2 methyltransferase MET-2 and can be recapitulated by a mutation in the putative H3K9me2 demethylase JHDM-1. These data suggest that lifespan is constrained by reduced H3K9me2 due to transcription-coupled H3K4me. wdr-5 mutants alleviate this burden, extending lifespan and enabling the inheritance of increased lifespan. Thus, H3K9me2 functions in the epigenetic establishment and inheritance of a complex trait. Based on this model, we propose that lifespan is limited by the germline in part because germline transcription reduces heterochromatin.


Sign in / Sign up

Export Citation Format

Share Document