Histone 3.3 Hotspot Mutations in Conventional Osteosarcomas

Total Page:16

File Type:pdf, Size:1020Kb

Histone 3.3 Hotspot Mutations in Conventional Osteosarcomas Koelsche et al. Clin Sarcoma Res (2017) 7:9 DOI 10.1186/s13569-017-0075-5 Clinical Sarcoma Research RESEARCH Open Access Histone 3.3 hotspot mutations in conventional osteosarcomas: a comprehensive clinical and molecular characterization of six H3F3A mutated cases Christian Koelsche1,2*† , Daniel Schrimpf1,2†, Lars Tharun3, Eva Roth4, Dominik Sturm4,5, David T. W. Jones4,5, Eva‑Kristin Renker6, Martin Sill7, Annika Baude8, Felix Sahm1,2, David Capper1,2, Melanie Bewerunge‑Hudler9, Wolfgang Hartmann10, Andreas E. Kulozik4, Iver Petersen11, Uta Flucke12, Hendrik W. B. Schreuder13, Reinhard Büttner3, Marc‑André Weber14, Peter Schirmacher15, Christoph Plass8, Stefan M. Pfster4,5, Andreas von Deimling1,2*‡ and Gunhild Mechtersheimer15*‡ Abstract Background: Histone 3.3 (H3.3) hotspot mutations in bone tumors occur in the vast majority of giant cell tumors of bone (GCTBs; 96%), chondroblastomas (95%) and in a few cases of osteosarcomas. However, clinical presentation, his‑ topathological features, and additional molecular characteristics of H3.3 mutant osteosarcomas are largely unknown. Methods: In this multicentre, retrospective study, a total of 106 conventional high-grade osteosarcomas, across all age groups were re-examined for hotspot mutations in the H3.3 coding genes H3F3A and H3F3B. H3.3 mutant osteo‑ sarcomas were re-evaluated in a multidisciplinary manner and analyzed for genome-wide DNA-methylation patterns and DNA copy number aberrations alongside H3.3 wild-type osteosarcomas and H3F3A G34W/L mutant GCTBs. Results: Six osteosarcomas (6/106) carried H3F3A hotspot mutations. No mutations were found in H3F3B. All patients with H3F3A mutant osteosarcoma were older than 30 years with a median age of 65 years. Copy number aberra‑ tions that are commonly encountered in high-grade osteosarcomas also occurred in H3F3A mutant osteosarcomas. Unlike a single osteosarcoma with a H3F3A K27M mutation, the DNA methylation profles of H3F3A G34W/R mutant osteosarcomas were clearly diferent from H3.3 wild-type osteosarcomas, but more closely related to GCTBs. The most diferentially methylated promoters between H3F3A G34W/R mutant and H3.3 wild-type osteosarcomas were in KLLN/ PTEN (p < 0.00005) and HIST1H2BB (p < 0.0005). *Correspondence: [email protected]‑heidelberg.de; [email protected]‑heidelberg.de; [email protected]‑heidelberg.de †Christian Koelsche and Daniel Schrimpf shared frst authorship ‡Andreas von Deimling and Gunhild Mechtersheimer shared last authorship 1 Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany 15 Department of General Pathology, Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany Full list of author information is available at the end of the article © The Author(s) 2017. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/ publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated. Koelsche et al. Clin Sarcoma Res (2017) 7:9 Page 2 of 11 Conclusions: H3.3 mutations in osteosarcomas may occur in H3F3A at mutational hotspots. They are overall rare, but become more frequent in osteosarcoma patients older than 30 years. Osteosarcomas carrying H3F3A G34W/R muta‑ tions are associated with epigenetic dysregulation of KLLN/PTEN and HIST1H2BB. Keywords: Mutation, H3.3, H3F3A, HIST1H2BB, KLLN, PTEN, Osteosarcoma, Giant cell tumor of bone, DNA methylation, Copy number alteration Background (α-thalassaemia/mental retardation syndrome X-linked) Osteosarcoma is a malignant bone forming neoplasm mutations, which result in a loss of its nuclear expression, with a broad spectrum of morphologies [1]. Conventional were found in osteosarcomas at a frequency of 20–30% high-grade osteosarcoma is the most common subtype [8, 16]. Loss of DAXX (death-domain associated protein) and typically arises in the long bones of the extremities expression has not yet been observed in any sarcoma with a predilection to the metaphysis, although any bone [17]. However, a high frequency of mutations in the two of the skeleton may be afected. Te incidence of osteo- genes encoding histone 3 variant 3 (H3.3) was revealed in sarcomas has a bimodal age distribution with a frst peak certain bone tumors. Giant cell tumors of bone (GCTBs) in the second decade of life and a second smaller peak in almost always carry mutations in H3F3A at codon 34, older adults [1, 2]. whereas chondroblastomas are characterized by a high Te etiology of osteosarcomas is still largely unknown. frequency of H3F3B mutations at codon 36 [18–20]. Lit- However, several genetic syndromes (e.g. Li-Fraumeni tle is known about H3.3 mutations in osteosarcomas. and hereditary retinoblastoma) greatly increase the Previous studies described single high-grade osteosarco- risk of developing osteosarcomas, especially in younger mas carrying H3.3 mutations, whereas other series did patients [3, 4]. Osteosarcomas in older patients often not. So the question arises whether osteosarcomas carry- develop from a precursor bone lesion that is prone for ing H3.3 mutations really exist or might rather be malig- malignant transformation (i.e. Paget disease) or may nant variants of GCTBs [8, 18–21]. occur after radiation treatment for other diseases, typi- Te present study addresses these issues by determin- cally after a latency period of several years [1, 5, 6]. ing the frequency of H3.3 mutations across the entire Te genomic landscape of osteosarcomas is com- age spectrum in de novo high-grade osteosarcomas of plex [7]. A remarkable attribute of primary and second- the conventional type. Genome-wide DNA methylation ary conventional osteosarcomas is their chromosomal and DNA copy number profling was applied to gain fur- instability leading to high aneuploidy [8]. Chromosomal ther molecular insight into osteosarcomas carrying H3.3 regions frequently afected by somatic structural vari- mutations. ations and copy number alterations often contain cell- cycle regulating genes such as TP53, RB1, or c-MYC [9, Methods 10]. Sequencing studies revealed recurrent mutations Tissues specimens afecting these genes, supporting their major role in Tissue samples of 106 de novo high-grade osteosarcomas osteosarcoma development [8]. However, the mutational of the conventional type (86 primary tumors, 3 recur- make-up of osteosarcomas is versatile, as some osteosar- rences, 17 metastases) and 14 GCTBs, one of them with comas also present with molecular signatures reminis- signs of malignancy in the local relapse two years after cent of BRCA1/2 defcient tumors [10]. Chromothripsis, surgical treatment, serving as H3.3 G34 mutant bone a single cellular catastrophic event resulting in extensive tumor control group were collected from the Institutes DNA rearrangements and DNA copy number changes, of Pathology of the University Hospital Heidelberg, has been observed in roughly one-third of osteosarcomas Cologne, Jena (Germany), and Nijemegen (the Neth- [11]. erlands). Patients’ characteristics are outlined in Addi- Alternative lengthening of telomeres (ALT), a tel- tional fle 1: Table S1. Tissue samples were banked in omere maintaining mechanism compensating progres- the archives of the Department of Pathology and admin- sive telomere attrition by homologous recombination istered by the tissue bank of the National Center for of telomere sequences, is associated with chromosomal Tumor Diseases (NCT), Heidelberg. Initial diagnoses instability in osteosarcomas [12]. Almost all osteosarco- were established by interdisciplinary oncology boards mas utilize ALT, highlighting its particular importance and were based on radiological fndings, standard histo- in these tumors [8, 13]. Somatic mutations in the H3.3- pathological criteria in conjunction with immunohisto- ATRX-DAXX chromatin-remodeling pathway have been chemical and molecular analyses according to the current linked to ALT in diferent tumor entities [14, 15]. ATRX WHO classifcation [1]. H3.3 mutant osteosarcomas were Koelsche et al. Clin Sarcoma Res (2017) 7:9 Page 3 of 11 correlated with clinical information and were histologi- Copy number profles were generated from the 450 k cally and radiologically re-evaluated where available. Te raw data using the ‘conumee’ R package and assessed local ethics committee approved these analyses. manually. Te dmpFinder method of the R package minf was used for the identifcation of diferentially methyl- DNA extraction and Sanger sequencing of H3F3A/B ated regions [22]. Both packages are available from the mutational hotspots Bioconductor repository (http://www.bioconductor.org). Source material for DNA extraction was formalin-fxed and parafn-embedded (FFPE) tumor tissue. Representa- Statistical analysis tive tumor tissue with highest available tumor content A two-sided t test was used to test for diferences was chosen for genomic DNA isolation, as microscopi- between the mean values for continuous variables. P val- cally documented on H&E stained parafn sections. ues of less than 0.05 were considered signifcant.
Recommended publications
  • Analysis of Trans Esnps Infers Regulatory Network Architecture
    Analysis of trans eSNPs infers regulatory network architecture Anat Kreimer Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2014 © 2014 Anat Kreimer All rights reserved ABSTRACT Analysis of trans eSNPs infers regulatory network architecture Anat Kreimer eSNPs are genetic variants associated with transcript expression levels. The characteristics of such variants highlight their importance and present a unique opportunity for studying gene regulation. eSNPs affect most genes and their cell type specificity can shed light on different processes that are activated in each cell. They can identify functional variants by connecting SNPs that are implicated in disease to a molecular mechanism. Examining eSNPs that are associated with distal genes can provide insights regarding the inference of regulatory networks but also presents challenges due to the high statistical burden of multiple testing. Such association studies allow: simultaneous investigation of many gene expression phenotypes without assuming any prior knowledge and identification of unknown regulators of gene expression while uncovering directionality. This thesis will focus on such distal eSNPs to map regulatory interactions between different loci and expose the architecture of the regulatory network defined by such interactions. We develop novel computational approaches and apply them to genetics-genomics data in human. We go beyond pairwise interactions to define network motifs, including regulatory modules and bi-fan structures, showing them to be prevalent in real data and exposing distinct attributes of such arrangements. We project eSNP associations onto a protein-protein interaction network to expose topological properties of eSNPs and their targets and highlight different modes of distal regulation.
    [Show full text]
  • Download Download
    Supplementary Figure S1. Results of flow cytometry analysis, performed to estimate CD34 positivity, after immunomagnetic separation in two different experiments. As monoclonal antibody for labeling the sample, the fluorescein isothiocyanate (FITC)- conjugated mouse anti-human CD34 MoAb (Mylteni) was used. Briefly, cell samples were incubated in the presence of the indicated MoAbs, at the proper dilution, in PBS containing 5% FCS and 1% Fc receptor (FcR) blocking reagent (Miltenyi) for 30 min at 4 C. Cells were then washed twice, resuspended with PBS and analyzed by a Coulter Epics XL (Coulter Electronics Inc., Hialeah, FL, USA) flow cytometer. only use Non-commercial 1 Supplementary Table S1. Complete list of the datasets used in this study and their sources. GEO Total samples Geo selected GEO accession of used Platform Reference series in series samples samples GSM142565 GSM142566 GSM142567 GSM142568 GSE6146 HG-U133A 14 8 - GSM142569 GSM142571 GSM142572 GSM142574 GSM51391 GSM51392 GSE2666 HG-U133A 36 4 1 GSM51393 GSM51394 only GSM321583 GSE12803 HG-U133A 20 3 GSM321584 2 GSM321585 use Promyelocytes_1 Promyelocytes_2 Promyelocytes_3 Promyelocytes_4 HG-U133A 8 8 3 GSE64282 Promyelocytes_5 Promyelocytes_6 Promyelocytes_7 Promyelocytes_8 Non-commercial 2 Supplementary Table S2. Chromosomal regions up-regulated in CD34+ samples as identified by the LAP procedure with the two-class statistics coded in the PREDA R package and an FDR threshold of 0.5. Functional enrichment analysis has been performed using DAVID (http://david.abcc.ncifcrf.gov/)
    [Show full text]
  • Histone H3.3 Maintains Genome Integrity During Mammalian Development
    Downloaded from genesdev.cshlp.org on September 25, 2021 - Published by Cold Spring Harbor Laboratory Press Histone H3.3 maintains genome integrity during mammalian development Chuan-Wei Jang, Yoichiro Shibata, Joshua Starmer, Della Yee, and Terry Magnuson Department of Genetics, Carolina Center for Genome Sciences, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599-7264, USA Histone H3.3 is a highly conserved histone H3 replacement variant in metazoans and has been implicated in many important biological processes, including cell differentiation and reprogramming. Germline and somatic mutations in H3.3 genomic incorporation pathway components or in H3.3 encoding genes have been associated with human congenital diseases and cancers, respectively. However, the role of H3.3 in mammalian development remains un- clear. To address this question, we generated H3.3-null mouse models through classical genetic approaches. We found that H3.3 plays an essential role in mouse development. Complete depletion of H3.3 leads to developmental retardation and early embryonic lethality. At the cellular level, H3.3 loss triggers cell cycle suppression and cell death. Surprisingly, H3.3 depletion does not dramatically disrupt gene regulation in the developing embryo. Instead, H3.3 depletion causes dysfunction of heterochromatin structures at telomeres, centromeres, and pericentromeric regions of chromosomes, leading to mitotic defects. The resulting karyotypical abnormalities and DNA damage lead to p53 pathway activation. In summary, our results reveal that an important function of H3.3 is to support chro- mosomal heterochromatic structures, thus maintaining genome integrity during mammalian development. [Keywords: histone H3.3; genome integrity; transcriptional regulation; cell proliferation; apoptosis; mouse embryonic development] Supplemental material is available for this article.
    [Show full text]
  • Integrating Single-Step GWAS and Bipartite Networks Reconstruction Provides Novel Insights Into Yearling Weight and Carcass Traits in Hanwoo Beef Cattle
    animals Article Integrating Single-Step GWAS and Bipartite Networks Reconstruction Provides Novel Insights into Yearling Weight and Carcass Traits in Hanwoo Beef Cattle Masoumeh Naserkheil 1 , Abolfazl Bahrami 1 , Deukhwan Lee 2,* and Hossein Mehrban 3 1 Department of Animal Science, University College of Agriculture and Natural Resources, University of Tehran, Karaj 77871-31587, Iran; [email protected] (M.N.); [email protected] (A.B.) 2 Department of Animal Life and Environment Sciences, Hankyong National University, Jungang-ro 327, Anseong-si, Gyeonggi-do 17579, Korea 3 Department of Animal Science, Shahrekord University, Shahrekord 88186-34141, Iran; [email protected] * Correspondence: [email protected]; Tel.: +82-31-670-5091 Received: 25 August 2020; Accepted: 6 October 2020; Published: 9 October 2020 Simple Summary: Hanwoo is an indigenous cattle breed in Korea and popular for meat production owing to its rapid growth and high-quality meat. Its yearling weight and carcass traits (backfat thickness, carcass weight, eye muscle area, and marbling score) are economically important for the selection of young and proven bulls. In recent decades, the advent of high throughput genotyping technologies has made it possible to perform genome-wide association studies (GWAS) for the detection of genomic regions associated with traits of economic interest in different species. In this study, we conducted a weighted single-step genome-wide association study which combines all genotypes, phenotypes and pedigree data in one step (ssGBLUP). It allows for the use of all SNPs simultaneously along with all phenotypes from genotyped and ungenotyped animals. Our results revealed 33 relevant genomic regions related to the traits of interest.
    [Show full text]
  • Genome-Wide Screen of Cell-Cycle Regulators in Normal and Tumor Cells
    bioRxiv preprint doi: https://doi.org/10.1101/060350; this version posted June 23, 2016. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Genome-wide screen of cell-cycle regulators in normal and tumor cells identifies a differential response to nucleosome depletion Maria Sokolova1, Mikko Turunen1, Oliver Mortusewicz3, Teemu Kivioja1, Patrick Herr3, Anna Vähärautio1, Mikael Björklund1, Minna Taipale2, Thomas Helleday3 and Jussi Taipale1,2,* 1Genome-Scale Biology Program, P.O. Box 63, FI-00014 University of Helsinki, Finland. 2Science for Life laboratory, Department of Biosciences and Nutrition, Karolinska Institutet, SE- 141 83 Stockholm, Sweden. 3Science for Life laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 21 Stockholm, Sweden To identify cell cycle regulators that enable cancer cells to replicate DNA and divide in an unrestricted manner, we performed a parallel genome-wide RNAi screen in normal and cancer cell lines. In addition to many shared regulators, we found that tumor and normal cells are differentially sensitive to loss of the histone genes transcriptional regulator CASP8AP2. In cancer cells, loss of CASP8AP2 leads to a failure to synthesize sufficient amount of histones in the S-phase of the cell cycle, resulting in slowing of individual replication forks. Despite this, DNA replication fails to arrest, and tumor cells progress in an elongated S-phase that lasts several days, finally resulting in death of most of the affected cells.
    [Show full text]
  • Whole Exome Sequencing in Families at High Risk for Hodgkin Lymphoma: Identification of a Predisposing Mutation in the KDR Gene
    Hodgkin Lymphoma SUPPLEMENTARY APPENDIX Whole exome sequencing in families at high risk for Hodgkin lymphoma: identification of a predisposing mutation in the KDR gene Melissa Rotunno, 1 Mary L. McMaster, 1 Joseph Boland, 2 Sara Bass, 2 Xijun Zhang, 2 Laurie Burdett, 2 Belynda Hicks, 2 Sarangan Ravichandran, 3 Brian T. Luke, 3 Meredith Yeager, 2 Laura Fontaine, 4 Paula L. Hyland, 1 Alisa M. Goldstein, 1 NCI DCEG Cancer Sequencing Working Group, NCI DCEG Cancer Genomics Research Laboratory, Stephen J. Chanock, 5 Neil E. Caporaso, 1 Margaret A. Tucker, 6 and Lynn R. Goldin 1 1Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; 2Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; 3Ad - vanced Biomedical Computing Center, Leidos Biomedical Research Inc.; Frederick National Laboratory for Cancer Research, Frederick, MD; 4Westat, Inc., Rockville MD; 5Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; and 6Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA ©2016 Ferrata Storti Foundation. This is an open-access paper. doi:10.3324/haematol.2015.135475 Received: August 19, 2015. Accepted: January 7, 2016. Pre-published: June 13, 2016. Correspondence: [email protected] Supplemental Author Information: NCI DCEG Cancer Sequencing Working Group: Mark H. Greene, Allan Hildesheim, Nan Hu, Maria Theresa Landi, Jennifer Loud, Phuong Mai, Lisa Mirabello, Lindsay Morton, Dilys Parry, Anand Pathak, Douglas R. Stewart, Philip R. Taylor, Geoffrey S. Tobias, Xiaohong R. Yang, Guoqin Yu NCI DCEG Cancer Genomics Research Laboratory: Salma Chowdhury, Michael Cullen, Casey Dagnall, Herbert Higson, Amy A.
    [Show full text]
  • Detection of H3K27M Mutation in Cases of Brain Stem Subependymoma
    Human Pathology (2019) 84,262–269 www.elsevier.com/locate/humpath Original contribution Detection of H3K27M mutation in cases of brain stem subependymoma☆ Kun Yao MD a,1, Zejun Duan MS a,1, Yin Wang MD b, Mingshan Zhang MD c, Tao Fan MD c, Bin Wu BS c, Xueling Qi BS a,⁎ aDepartment of Pathology, San Bo Brain Hospital, Capital Medical University, Haidian District, Beijing, China bDepartment of Pathology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China cDepartment of Neurosurgery, San Bo Brain Hospital, Capital Medical University, Haidian District, Beijing, China Received 5 August 2018; revised 2 October 2018; accepted 11 October 2018 Keywords: Summary Subependymomas are rare, slow-growing, grade I glial tumors of the central nervous system. Re- H3K27M mutation; cently, diffuse midline gliomas with mutations in the H3.1 or H3.3 genes at the position of amino acid 27, Subependymoma; resulting in the replacement of lysine by methionine (K27M), were defined as the new grade IV entity. As Brain stem neoplasm; H3K27M mutations have been reported in midline gliomas, gangliogliomas, and pilocytic astrocytomas, Sanger sequencing whether they occur in midline subependymomas has been unclear. We determined whether any such muta- tions can be found in them and analyzed the prognostic relevance of any such mutations in subependymo- mas. Four subependymomas, all in the brain stem, harbored H3K27M mutations. No such mutation was found in any of the subependymomas from other locations. The mutations were identified by immunohisto- chemical stains and confirmed with Sanger sequencing. The median follow-up of the patients with the mutations in their tumors was 3.2 years, and 3 are still alive, having received no adjuvant therapy.
    [Show full text]
  • "The Genecards Suite: from Gene Data Mining to Disease Genome Sequence Analyses". In: Current Protocols in Bioinformat
    The GeneCards Suite: From Gene Data UNIT 1.30 Mining to Disease Genome Sequence Analyses Gil Stelzer,1,5 Naomi Rosen,1,5 Inbar Plaschkes,1,2 Shahar Zimmerman,1 Michal Twik,1 Simon Fishilevich,1 Tsippi Iny Stein,1 Ron Nudel,1 Iris Lieder,2 Yaron Mazor,2 Sergey Kaplan,2 Dvir Dahary,2,4 David Warshawsky,3 Yaron Guan-Golan,3 Asher Kohn,3 Noa Rappaport,1 Marilyn Safran,1 and Doron Lancet1,6 1Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel 2LifeMap Sciences Ltd., Tel Aviv, Israel 3LifeMap Sciences Inc., Marshfield, Massachusetts 4Toldot Genetics Ltd., Hod Hasharon, Israel 5These authors contributed equally to the paper 6Corresponding author GeneCards, the human gene compendium, enables researchers to effectively navigate and inter-relate the wide universe of human genes, diseases, variants, proteins, cells, and biological pathways. Our recently launched Version 4 has a revamped infrastructure facilitating faster data updates, better-targeted data queries, and friendlier user experience. It also provides a stronger foundation for the GeneCards suite of companion databases and analysis tools. Improved data unification includes gene-disease links via MalaCards and merged biological pathways via PathCards, as well as drug information and proteome expression. VarElect, another suite member, is a phenotype prioritizer for next-generation sequencing, leveraging the GeneCards and MalaCards knowledgebase. It au- tomatically infers direct and indirect scored associations between hundreds or even thousands of variant-containing genes and disease phenotype terms. Var- Elect’s capabilities, either independently or within TGex, our comprehensive variant analysis pipeline, help prepare for the challenge of clinical projects that involve thousands of exome/genome NGS analyses.
    [Show full text]
  • Effects of H3.3G34V Mutation on Genomic H3K36 and H3K27 Methylation Patterns in Isogenic Pediatric Glioma Cells
    Huang et al. acta neuropathol commun (2020) 8:219 https://doi.org/10.1186/s40478-020-01092-4 RESEARCH Open Access Efects of H3.3G34V mutation on genomic H3K36 and H3K27 methylation patterns in isogenic pediatric glioma cells Tina Yi‑Ting Huang1, Andrea Piunti2, Jin Qi1, Marc Morgan2, Elizabeth Bartom2, Ali Shilatifard2 and Amanda M. Saratsis1,2,3* Abstract Histone H3.3 mutation (H3F3A) occurs in 50% of cortical pediatric high‑grade gliomas. This mutation replaces glycine 34 with arginine or valine (G34R/V), impairing SETD2 activity (H3K36‑specifc trimethyltransferase). Consequently, reduced H3K36me3 is observed on H3.3G34V nucleosomes relative to wild‑type, contributing to genomic instabil‑ ity and driving a distinct gene expression signature associated with tumorigenesis. However, it is not known if this diferential H3K36me3 enrichment is due to H3.3G34V mutant protein alone. Therefore, we set to elucidate the efect of H3.3G34V mutant protein in pediatric glioma on H3K36me3, H3K27me3 and H3.3 enrichment in vitro. We found that the doxycycline‑inducible shRNA knockdown of mutant H3F3A encoding the H3.3G34V protein resulted in loss of H3.3G34V enrichment and increased H3K36me3 enrichment throughout the genome. After knockdown, H3.3G34V enrichment was preserved at loci observed to have the greatest H3.3G34V and H3K36me3 enrichment prior to knockdown. Induced expression of mutant H3.3G34V protein in vitro was insufcient to induce genomic H3K36me3 enrichment patterns observed in H3.3G34V mutant glioma cells. We also observed strong co‑enrichment of H3.3G34V and wild‑type H3.3 protein, as well as greater H3K27me3 enrichment, in cells expressing H3.3G34V.
    [Show full text]
  • Histone H3F3A and HIST1H3B K27M Mutations Define Two Subgroups of Diffuse Intrinsic Pontine Gliomas with Different Prognosis and Phenotypes
    Acta Neuropathol (2015) 130:815–827 DOI 10.1007/s00401-015-1478-0 ORIGINAL PAPER Histone H3F3A and HIST1H3B K27M mutations define two subgroups of diffuse intrinsic pontine gliomas with different prognosis and phenotypes David Castel1,2 · Cathy Philippe1 · Raphaël Calmon3 · Ludivine Le Dret1 · Nathalène Truffaux1 · Nathalie Boddaert3 · Mélanie Pagès7 · Kathryn R. Taylor4 · Patrick Saulnier5 · Ludovic Lacroix5 · Alan Mackay4 · Chris Jones4 · Christian Sainte‑Rose6 · Thomas Blauwblomme6 · Felipe Andreiuolo7 · Stephanie Puget6 · Jacques Grill1,2 · Pascale Varlet7 · Marie‑Anne Debily1,8 Received: 24 June 2015 / Revised: 8 September 2015 / Accepted: 10 September 2015 / Published online: 23 September 2015 © The Author(s) 2015. This article is published with open access at Springerlink.com Abstract Diffuse intrinsic pontine glioma (DIPG) is the a systematic stereotactic biopsy and were included in most severe paediatric solid tumour, with no significant this observational retrospective study. Histone H3 genes therapeutic progress made in the past 50 years. Recent mutations were assessed by immunochemistry and direct studies suggest that diffuse midline glioma, H3-K27M sequencing, whilst global gene expression profiling and mutant, may comprise more than one biological entity. chromosomal imbalances were determined by microar- The aim of the study was to determine the clinical and bio- rays. A full description of the MRI findings at diagnosis logical variables that most impact their prognosis. Ninety- and at relapse was integrated with the molecular profiling one patients with classically defined DIPG underwent data and clinical outcome. All DIPG but one were found to harbour either a somatic H3-K27M mutation and/or loss of H3K27 trimethylation. We also discovered a novel K27M This work was presented at the International Symposium mutation in HIST2H3C, and a lysine-to-isoleucine substitu- of Pediatric Neuro-Oncology meeting held in June 2014 in tion (K27I) in H3F3A, also creating a loss of trimethyla- Singapore.
    [Show full text]
  • Datasheet: VPA00629KT Product Details
    Datasheet: VPA00629KT Description: HISTONE H3F3A ANTIBODY WITH CONTROL LYSATE Specificity: HISTONE H3F3A Format: Purified Product Type: PrecisionAb™ Polyclonal Isotype: Polyclonal IgG Quantity: 2 Westerns Product Details Applications This product has been reported to work in the following applications. This information is derived from testing within our laboratories, peer-reviewed publications or personal communications from the originators. Please refer to references indicated for further information. For general protocol recommendations, please visit www.bio-rad-antibodies.com/protocols. Yes No Not Determined Suggested Dilution Western Blotting 1/1000 PrecisionAb antibodies have been extensively validated for the western blot application. The antibody has been validated at the suggested dilution. Where this product has not been tested for use in a particular technique this does not necessarily exclude its use in such procedures. Further optimization may be required dependant on sample type. Target Species Human Species Cross Reacts with: Mouse Reactivity N.B. Antibody reactivity and working conditions may vary between species. Product Form Purified IgG - liquid Preparation 20μl Rabbit polyclonal antibody purified by affinity chromatography Buffer Solution Phosphate buffered saline Preservative 0.09% Sodium Azide Stabilisers 1% Bovine Serum Albumin Immunogen Recombinant histone H3F3A protein External Database Links UniProt: P84243 Related reagents Entrez Gene: 3021 H3F3B Related reagents Synonyms H3.3A, H3.3B, H3F3 Page 1 of 3 Specificity Rabbit anti Human histone H3F3A antibody recognizes histone H3.3. Histones are basic nuclear proteins that are responsible for the nucleosome structure of the chromosomal fiber in eukaryotes. Two molecules of each of the four core histones (H2A, H2B, H3, and H4) form an octamer, around which approximately 146 bp of DNA is wrapped in repeating units, called nucleosomes.
    [Show full text]
  • Germline Mutations in Histone 3 Family 3A and 3B Cause a Previously Unidentified Neurodegenerative Disorder in 46 Patients
    Washington University School of Medicine Digital Commons@Becker Open Access Publications 2020 Histone H3.3 beyond cancer: Germline mutations in Histone 3 Family 3A and 3B cause a previously unidentified neurodegenerative disorder in 46 patients Laura Bryant Marcia C Willing Linda Manwaring et al Follow this and additional works at: https://digitalcommons.wustl.edu/open_access_pubs SCIENCE ADVANCES | RESEARCH ARTICLE GENETICS Copyright © 2020 The Authors, some rights reserved; Histone H3.3 beyond cancer: Germline mutations in exclusive licensee American Association Histone 3 Family 3A and 3B cause a previously for the Advancement of Science. No claim to unidentified neurodegenerative disorder in 46 patients original U.S. Government Laura Bryant1*, Dong Li1*, Samuel G. Cox2, Dylan Marchione3, Evan F. Joiner4, Khadija Wilson3, Works. Distributed 3 5 1 1 6 6 under a Creative Kevin Janssen , Pearl Lee , Michael E. March , Divya Nair , Elliott Sherr , Brieana Fregeau , Commons Attribution 7 7 8 9 Klaas J. Wierenga , Alexandrea Wadley , Grazia M. S. Mancini , Nina Powell-Hamilton , NonCommercial 10 11 12 12 13 Jiddeke van de Kamp , Theresa Grebe , John Dean , Alison Ross , Heather P. Crawford , License 4.0 (CC BY-NC). Zoe Powis14, Megan T. Cho15, Marcia C. Willing16, Linda Manwaring16, Rachel Schot8, Caroline Nava17,18, Alexandra Afenjar19, Davor Lessel20,21, Matias Wagner22,23,24, Thomas Klopstock25,26,27, Juliane Winkelmann22,24,27,28, Claudia B. Catarino25, Kyle Retterer15, Jane L. Schuette29, Jeffrey W. Innis29, Amy Pizzino30,31, Sabine Lüttgen32, Jonas Denecke32, 22,24 15 3 30,31 Tim M. Strom , Kristin G. Monaghan ; DDD Study, Zuo-Fei Yuan , Holly Dubbs , Downloaded from Renee Bend33, Jennifer A.
    [Show full text]