25 results on '"Jon A. Oyer"'
Search Results
2. Supplementary Table 4: H4 from A Mutation in Histone H2B Represents a New Class of Oncogenic Driver
- Author
-
Jonathan D. Licht, Jan Liphardt, Alexander J. Ruthenburg, Neil L. Kelleher, George C. Schatz, Tanmay P. Lele, Alberto Riva, Qiao Zhang, Tao Yu, Adrian T. Grzybowski, Rajarshi P. Ghosh, Xiaoxiao Huang, Jon A. Oyer, Behnam Nabet, Christine M. Will, Eliza C. Small, Aditya Bele, and Richard L. Bennett
- Abstract
List of all H4 missense mutations in cBioPortal nad dbSNP analysis
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Table 2: H2B from A Mutation in Histone H2B Represents a New Class of Oncogenic Driver
- Author
-
Jonathan D. Licht, Jan Liphardt, Alexander J. Ruthenburg, Neil L. Kelleher, George C. Schatz, Tanmay P. Lele, Alberto Riva, Qiao Zhang, Tao Yu, Adrian T. Grzybowski, Rajarshi P. Ghosh, Xiaoxiao Huang, Jon A. Oyer, Behnam Nabet, Christine M. Will, Eliza C. Small, Aditya Bele, and Richard L. Bennett
- Abstract
List of all H2B missense mutations in cBioPortal and dbSNP analysis
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Table 3: H3 from A Mutation in Histone H2B Represents a New Class of Oncogenic Driver
- Author
-
Jonathan D. Licht, Jan Liphardt, Alexander J. Ruthenburg, Neil L. Kelleher, George C. Schatz, Tanmay P. Lele, Alberto Riva, Qiao Zhang, Tao Yu, Adrian T. Grzybowski, Rajarshi P. Ghosh, Xiaoxiao Huang, Jon A. Oyer, Behnam Nabet, Christine M. Will, Eliza C. Small, Aditya Bele, and Richard L. Bennett
- Abstract
List of all H3 missense mutations in cBioPortal and dbSNP analysis
- Published
- 2023
- Full Text
- View/download PDF
5. Supplemental Text and Figures from A Mutation in Histone H2B Represents a New Class of Oncogenic Driver
- Author
-
Jonathan D. Licht, Jan Liphardt, Alexander J. Ruthenburg, Neil L. Kelleher, George C. Schatz, Tanmay P. Lele, Alberto Riva, Qiao Zhang, Tao Yu, Adrian T. Grzybowski, Rajarshi P. Ghosh, Xiaoxiao Huang, Jon A. Oyer, Behnam Nabet, Christine M. Will, Eliza C. Small, Aditya Bele, and Richard L. Bennett
- Abstract
Supplemental materials and methods and Supplemental Figures 1-7
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Table 1: H2A from A Mutation in Histone H2B Represents a New Class of Oncogenic Driver
- Author
-
Jonathan D. Licht, Jan Liphardt, Alexander J. Ruthenburg, Neil L. Kelleher, George C. Schatz, Tanmay P. Lele, Alberto Riva, Qiao Zhang, Tao Yu, Adrian T. Grzybowski, Rajarshi P. Ghosh, Xiaoxiao Huang, Jon A. Oyer, Behnam Nabet, Christine M. Will, Eliza C. Small, Aditya Bele, and Richard L. Bennett
- Abstract
List of all H2A missense mutations in cBioPortal and dbSNP analysis
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Methods and Figures from PRC2 Inhibitors Overcome Glucocorticoid Resistance Driven by NSD2 Mutation in Pediatric Acute Lymphoblastic Leukemia
- Author
-
Jonathan D. Licht, Richard B. Lock, Matthew D. Hall, Adolfo A. Ferrando, Richard L. Bennett, Alberto Riva, Min Shen, Christine M. Will, Jon A. Oyer, Marta Kulis, Alok Swaroop, Catalina Troche, Heidi L. Casellas Román, Duohui Jing, Jacob S. Roth, Daphne Dupéré-Richér, Crissandra Piper, Jonathan H. Shrimp, Julia Hlavka-Zhang, and Jianping Li
- Abstract
Supplementary Methods and Figures
- Published
- 2023
- Full Text
- View/download PDF
8. Data from PRC2 Inhibitors Overcome Glucocorticoid Resistance Driven by NSD2 Mutation in Pediatric Acute Lymphoblastic Leukemia
- Author
-
Jonathan D. Licht, Richard B. Lock, Matthew D. Hall, Adolfo A. Ferrando, Richard L. Bennett, Alberto Riva, Min Shen, Christine M. Will, Jon A. Oyer, Marta Kulis, Alok Swaroop, Catalina Troche, Heidi L. Casellas Román, Duohui Jing, Jacob S. Roth, Daphne Dupéré-Richér, Crissandra Piper, Jonathan H. Shrimp, Julia Hlavka-Zhang, and Jianping Li
- Abstract
Mutations in epigenetic regulators are common in relapsed pediatric acute lymphoblastic leukemia (ALL). Here, we uncovered the mechanism underlying the relapse of ALL driven by an activating mutation of the NSD2 histone methyltransferase (p.E1099K). Using high-throughput drug screening, we found that NSD2-mutant cells were specifically resistant to glucocorticoids. Correction of this mutation restored glucocorticoid sensitivity. The transcriptional response to glucocorticoids was blocked in NSD2-mutant cells due to depressed glucocorticoid receptor (GR) levels and the failure of glucocorticoids to autoactivate GR expression. Although H3K27me3 was globally decreased by NSD2 p.E1099K, H3K27me3 accumulated at the NR3C1 (GR) promoter. Pretreatment of NSD2 p.E1099K cell lines and patient-derived xenograft samples with PRC2 inhibitors reversed glucocorticoid resistance in vitro and in vivo. PRC2 inhibitors restored NR3C1 autoactivation by glucocorticoids, increasing GR levels and allowing GR binding and activation of proapoptotic genes. These findings suggest a new therapeutic approach to relapsed ALL associated with NSD2 mutation.Significance:NSD2 histone methyltransferase mutations observed in relapsed pediatric ALL drove glucocorticoid resistance by repression of the GR and abrogation of GR gene autoactivation due to accumulation of K3K27me3 at its promoter. Pretreatment with PRC2 inhibitors reversed resistance, suggesting a new therapeutic approach to these patients with ALL.This article is highlighted in the In This Issue feature, p. 1
- Published
- 2023
- Full Text
- View/download PDF
9. Data from A Mutation in Histone H2B Represents a New Class of Oncogenic Driver
- Author
-
Jonathan D. Licht, Jan Liphardt, Alexander J. Ruthenburg, Neil L. Kelleher, George C. Schatz, Tanmay P. Lele, Alberto Riva, Qiao Zhang, Tao Yu, Adrian T. Grzybowski, Rajarshi P. Ghosh, Xiaoxiao Huang, Jon A. Oyer, Behnam Nabet, Christine M. Will, Eliza C. Small, Aditya Bele, and Richard L. Bennett
- Abstract
By examination of the cancer genomics database, we identified a new set of mutations in core histones that frequently recur in cancer patient samples and are predicted to disrupt nucleosome stability. In support of this idea, we characterized a glutamate to lysine mutation of histone H2B at amino acid 76 (H2B-E76K), found particularly in bladder and head and neck cancers, that disrupts the interaction between H2B and H4. Although H2B-E76K forms dimers with H2A, it does not form stable histone octamers with H3 and H4 in vitro, and when reconstituted with DNA forms unstable nucleosomes with increased sensitivity to nuclease. Expression of the equivalent H2B mutant in yeast restricted growth at high temperature and led to defective nucleosome-mediated gene repression. Significantly, H2B-E76K expression in the normal mammary epithelial cell line MCF10A increased cellular proliferation, cooperated with mutant PIK3CA to promote colony formation, and caused a significant drift in gene expression and fundamental changes in chromatin accessibility, particularly at gene regulatory elements. Taken together, these data demonstrate that mutations in the globular domains of core histones may give rise to an oncogenic program due to nucleosome dysfunction and deregulation of gene expression.Significance:Mutations in the core histones frequently occur in cancer and represent a new mechanism of epigenetic dysfunction that involves destabilization of the nucleosome, deregulation of chromatin accessibility, and alteration of gene expression to drive cellular transformation.See related commentary by Sarthy and Henikoff, p. 1346.This article is highlighted in the In This Issue feature, p. 1325
- Published
- 2023
- Full Text
- View/download PDF
10. PRC2 Inhibitors Overcome Glucocorticoid Resistance Driven by NSD2 Mutation in Pediatric Acute Lymphoblastic Leukemia
- Author
-
Crissandra Piper, Matthew D. Hall, Marta Kulis, Richard L. Bennett, Alok Swaroop, Min Shen, Richard B. Lock, Jonathan H. Shrimp, Jon A. Oyer, Christine Will, Alberto Riva, Heidi L. Casellas Roman, Duohui Jing, Jianping Li, Catalina Troche, Adolfo A. Ferrando, Jacob S. Roth, Daphné Dupéré-Richer, Jonathan D. Licht, and Julia Cathryn Hlavka-Zhang
- Subjects
Mutation ,biology ,business.industry ,medicine.disease_cause ,Glucocorticoid receptor ,Glucocorticoid Sensitivity ,Oncology ,In vivo ,Cell culture ,Histone methyltransferase ,medicine ,Cancer research ,biology.protein ,Epigenetics ,PRC2 ,business ,hormones, hormone substitutes, and hormone antagonists - Abstract
Mutations in epigenetic regulators are common in relapsed pediatric acute lymphoblastic leukemia (ALL). Here, we uncovered the mechanism underlying the relapse of ALL driven by an activating mutation of the NSD2 histone methyltransferase (p.E1099K). Using high-throughput drug screening, we found that NSD2-mutant cells were specifically resistant to glucocorticoids. Correction of this mutation restored glucocorticoid sensitivity. The transcriptional response to glucocorticoids was blocked in NSD2-mutant cells due to depressed glucocorticoid receptor (GR) levels and the failure of glucocorticoids to autoactivate GR expression. Although H3K27me3 was globally decreased by NSD2 p.E1099K, H3K27me3 accumulated at the NR3C1 (GR) promoter. Pretreatment of NSD2 p.E1099K cell lines and patient-derived xenograft samples with PRC2 inhibitors reversed glucocorticoid resistance in vitro and in vivo. PRC2 inhibitors restored NR3C1 autoactivation by glucocorticoids, increasing GR levels and allowing GR binding and activation of proapoptotic genes. These findings suggest a new therapeutic approach to relapsed ALL associated with NSD2 mutation. Significance: NSD2 histone methyltransferase mutations observed in relapsed pediatric ALL drove glucocorticoid resistance by repression of the GR and abrogation of GR gene autoactivation due to accumulation of K3K27me3 at its promoter. Pretreatment with PRC2 inhibitors reversed resistance, suggesting a new therapeutic approach to these patients with ALL. This article is highlighted in the In This Issue feature, p. 1
- Published
- 2022
- Full Text
- View/download PDF
11. Polycomb- and REST-associated histone deacetylases are independent pathways toward a mature neuronal phenotype
- Author
-
James C McGann, Jon A Oyer, Saurabh Garg, Huilan Yao, Jun Liu, Xin Feng, Lujian Liao, John R Yates III, and Gail Mandel
- Subjects
ES cell ,REST ,Polycomb ,poised ,histone deacetylase ,neuronal ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The bivalent hypothesis posits that genes encoding developmental regulators required for early lineage decisions are poised in stem/progenitor cells by the balance between a repressor histone modification (H3K27me3), mediated by the Polycomb Repressor Complex 2 (PRC2), and an activator modification (H3K4me3). In this study, we test whether this mechanism applies equally to genes that are not required until terminal differentiation. We focus on the RE1 Silencing Transcription Factor (REST) because it is expressed highly in stem cells and is an established global repressor of terminal neuronal genes. Elucidation of the REST complex, and comparison of chromatin marks and gene expression levels in control and REST-deficient stem cells, shows that REST target genes are poised by a mechanism independent of Polycomb, even at promoters which bear the H3K27me3 mark. Specifically, genes under REST control are actively repressed in stem cells by a balance of the H3K4me3 mark and a repressor complex that relies on histone deacetylase activity. Thus, chromatin distinctions between pro-neural and terminal neuronal genes are established at the embryonic stem cell stage by two parallel, but distinct, repressor pathways.
- Published
- 2014
- Full Text
- View/download PDF
12. PRC2 Inhibitors Overcome Glucocorticoid Resistance Driven by
- Author
-
Jianping, Li, Julia, Hlavka-Zhang, Jonathan H, Shrimp, Crissandra, Piper, Daphne, Dupéré-Richér, Jacob S, Roth, Duohui, Jing, Heidi L, Casellas Román, Catalina, Troche, Alok, Swaroop, Marta, Kulis, Jon A, Oyer, Christine M, Will, Min, Shen, Alberto, Riva, Richard L, Bennett, Adolfo A, Ferrando, Matthew D, Hall, Richard B, Lock, and Jonathan D, Licht
- Subjects
Male ,Cell Survival ,Histone-Lysine N-Methyltransferase ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Repressor Proteins ,Drug Resistance, Neoplasm ,Cell Line, Tumor ,Mutation ,Histone Methyltransferases ,Humans ,Female ,Enzyme Inhibitors ,Child ,Glucocorticoids - Abstract
Mutations in epigenetic regulators are common in relapsed pediatric acute lymphoblastic leukemia (ALL). Here, we uncovered the mechanism underlying the relapse of ALL driven by an activating mutation of the
- Published
- 2020
13. Aberrant epigenetic silencing is triggered by a transient reduction in gene expression.
- Author
-
Jon A Oyer, Adrian Chu, Sukhmani Brar, and Mitchell S Turker
- Subjects
Medicine ,Science - Abstract
Aberrant epigenetic silencing plays a major role in cancer formation by inactivating tumor suppressor genes. While the endpoints of aberrant silencing are known, i.e., promoter region DNA methylation and altered histone modifications, the triggers of silencing are not known. We used the tet-off system to test the hypothesis that a transient reduction in gene expression will sensitize a promoter to undergo epigenetic silencing.The tet responsive promoter (P(TRE)) was used to drive expression of the selectable human HPRT cDNA in independent transfectants of an Hprt deficient mouse cell line. In this system, high basal HPRT expression is greatly reduced when doxycycline (Dox) is added to the culture medium. Exposure of the P(TRE)-HPRT transfectants to Dox induced HPRT deficient clones in a time dependent manner. A molecular analysis demonstrated promoter region DNA methylation, loss of histone modifications associated with expression (i.e., H3 lysine 9 and 14 acetylation and lysine 4 methylation), and acquisition of the repressive histone modification H3 lysine 9 methylation. These changes, which are consistent with aberrant epigenetic silencing, were not present in the Dox-treated cultures, with the exception of reduced H3 lysine 14 acetylation. Silenced alleles readily reactivated spontaneously or after treatment of cells with inhibitors of histone deacetylation and/or DNA methylation, but re-silencing of reactivated alleles did not require a new round of Dox exposure. Inhibition of histone deacetylation inhibited both the induction of silencing and re-silencing, whereas inhibition of DNA methylation had no such effect.This study demonstrates that a transient reduction in gene expression triggers a pathway for aberrant silencing in mammalian cells and identifies histone deacetylation as a critical early step in this process. DNA methylation, in contrast, is a secondary step in the silencing pathway under study. A model to explain these observations is offered.
- Published
- 2009
- Full Text
- View/download PDF
14. NSD2-E1099K Mutation Leads to Glucocorticoid-Resistant B Cell Lymphocytic Leukemia in Mice
- Author
-
Daphné Dupéré-Richer, Jon A. Oyer, Jonathan D. Licht, Heidi L. Casellas Roman, Alberto Riva, Christine Will, Richard L. Bennett, Alok Swaroop, Jianping Li, Catalina Troche, and Crissandra Piper
- Subjects
B cell lymphocytic leukemia ,Immunology ,Mutation (genetic algorithm) ,medicine ,Cancer research ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Glucocorticoid ,medicine.drug - Abstract
Background: NSD2 (nuclear receptor binding SET domain protein 2) is a histone methyltransferase specific for dimethylation of histone H3 lysine 36 (H3K36me2), a modification associated with gene activation. In pediatric acute lymphoblastic leukemia (ALL), particularly at relapse, a gain of function mutation (E1099K) of NSD2 is found in 10-15% of cases. The NSD2 mutation is found in addition to fusion proteins such as E2A-PBX and ETV6-RUNX1. The mutation can be subclonal at diagnosis and dominant at relapse, suggesting a link to therapeutic resistance. The NSD2-E1099K mutation affects gene expression through an increase in H3K36me2 and a decrease in H3K27me3. Using CRISPR/Cas9-edited isogenic ALL cell lines, we found that NSD2-E1099K mutation drove oncogenic programming by altering chromatin architecture, gene expression and enhancing cell growth, migration and infiltration to the central neural system (CNS). NSD2 mutation caused resistance of ALL cells to glucocorticoids (GC) by blocking genome wide binding of the glucocorticoid receptor (GR, encoded by NR3C1 gene) preventing GC-mediated induction of pro-apoptotic genes. NR3C1 levels were depressed in NSD2-E1099K cells and GC failed to induce autoactivation of NR3C1. While H3K27me3 was globally decreased by NSD2-E1099K, increased H3K27me3 was noted at the promoter of NR3C1, suggesting a novel role of polycomb repressive complex 2 as a therapeutic target for relapsed ALL with NSD2 mutation. While NSD2 is highly expressed in B cells and NSD2 knockout causes defects in B cell development, how the NSD2 mutation affects B cell development and leukemia occurrence in vivo is uncertain. Aims: To determine the role of NSD2 mutation in the pathogenesis of lymphocytic malignancies and GC resistance in a mouse model. Methods: We generated a conditional NSD2-E1099K knock-in mouse model in which the NSD2-E1099K allele was placed in the Rosa26 locus and expressed in B cells under the control of Cd19-Cre (Cd19+/-NSD2E1099K/WT). The resulting phenotype was characterized through peripheral blood counts, cellular morphology and histology of blood smears, bone marrow (BM), spleen and liver, flow cytometric analysis, germinal center B cells (GCB) immunization, BM transplantation, and hematopoiesis analysis in a CD3-/- background. We further established mouse leukemia cell lines with NSD2 mutation for functional analysis. RNA-Seq, real time PCR, immunoblotting, and apoptosis analysis (Annexin V/PI staining) following GC treatment were performed to demonstrate the effects of NSD2 mutation on histone modifications, transcriptome and GC resistance. Results: The NSD2-E1099K mutation increased H3K36me2 and decreased H3K27me3 in isolated B cells from mouse BM and spleen. Mice were aged and did not develop signs of malignancy and RNA-sequencing showed few differences between B cells with or without the NSD2 mutation. However, after immunizing the mice with sheep red blood cells (SRBC), more GCBs were seen in the spleen of NSD2 mutant mice, suggesting mutant NSD2 stimulated germinal center hyperplasia. Transplantation of BM cells from mice expressing NSD2-E1099K into lethally irradiated recipients lead to an expansion of B cells while myeloid and T cells and life span of the recipients impaired. The NSD2 knock-in mouse model was crossed with Cd3-/- mice to create Cd19+/-Cd3-/-NSD2E1099K/WT mice, which within 2 months of birth developed a disease resembling an immature B lymphocytic leukemia (B220+CD19+IgM+IgD-CD5-) with infiltration of the spleen, liver and CNS and a median survival of 4.8 months. These tumors could be transplanted into immunodeficient mice but not immunocompetent mice. RNA seq analysis of these cells revealed 6,815 genes (3,295 upregulated and 3,520 downregulated) differentially expressed in NSD2 mutant B cells compared to normal B cells. The upregulated genes were related to abnormal immunoglobulin level , B cell activation, T-helper 1 physiology, and decreased B cell apoptosis. Importantly, the NSD2 mutant leukemic cells displayed depressed level of NR3C1 gene expression and GC resistance. Conclusions: The NSD2 mutation alters B cell development, particularly in an immunodeficient background and causes B cells to become resistant to glucocorticoids. The inability of the mutation to generate disease on its own except in an immunodeficient background suggests genes that collaborate with NSD2 in ALL may play a role in immune escape. Disclosures No relevant conflicts of interest to declare.
- Published
- 2020
- Full Text
- View/download PDF
15. A Mutation in Histone H2B Represents a New Class of Oncogenic Driver
- Author
-
Jan Liphardt, Jon A. Oyer, Adrian T. Grzybowski, Rajarshi P. Ghosh, Alberto Riva, Richard L. Bennett, Qiao Zhang, Neil L. Kelleher, Aditya Bele, Jonathan D. Licht, Tanmay P. Lele, Behnam Nabet, Eliza C. Small, Tao Yu, Alexander J. Ruthenburg, George C. Schatz, Xiaoxiao Huang, and Christine Will
- Subjects
0301 basic medicine ,Mutant ,Mutation, Missense ,Gene Expression ,medicine.disease_cause ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,Yeasts ,medicine ,Histone H2B ,Nucleosome ,Humans ,Epigenetics ,Alleles ,Mutation ,biology ,Gene Expression Profiling ,Oncogenes ,Cell biology ,Chromatin ,Nucleosomes ,Gene expression profiling ,030104 developmental biology ,Histone ,Cell Transformation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Protein Multimerization - Abstract
By examination of the cancer genomics database, we identified a new set of mutations in core histones that frequently recur in cancer patient samples and are predicted to disrupt nucleosome stability. In support of this idea, we characterized a glutamate to lysine mutation of histone H2B at amino acid 76 (H2B-E76K), found particularly in bladder and head and neck cancers, that disrupts the interaction between H2B and H4. Although H2B-E76K forms dimers with H2A, it does not form stable histone octamers with H3 and H4 in vitro, and when reconstituted with DNA forms unstable nucleosomes with increased sensitivity to nuclease. Expression of the equivalent H2B mutant in yeast restricted growth at high temperature and led to defective nucleosome-mediated gene repression. Significantly, H2B-E76K expression in the normal mammary epithelial cell line MCF10A increased cellular proliferation, cooperated with mutant PIK3CA to promote colony formation, and caused a significant drift in gene expression and fundamental changes in chromatin accessibility, particularly at gene regulatory elements. Taken together, these data demonstrate that mutations in the globular domains of core histones may give rise to an oncogenic program due to nucleosome dysfunction and deregulation of gene expression. Significance: Mutations in the core histones frequently occur in cancer and represent a new mechanism of epigenetic dysfunction that involves destabilization of the nucleosome, deregulation of chromatin accessibility, and alteration of gene expression to drive cellular transformation. See related commentary by Sarthy and Henikoff, p. 1346. This article is highlighted in the In This Issue feature, p. 1325
- Published
- 2019
16. Emerging Approaches for the Identification of Protein Targets of Small Molecules - A Practitioners' Perspective
- Author
-
Anil Vasudevan, Paul L. Richardson, Jon A. Oyer, Scott E. Warder, Kenneth M. Comess, Henning Stöckmann, and Shaun M. McLoughlin
- Subjects
0301 basic medicine ,Proteome ,010405 organic chemistry ,Drug discovery ,Chemistry ,Phenotypic screening ,Data complexity ,01 natural sciences ,Data science ,0104 chemical sciences ,High-Throughput Screening Assays ,Small Molecule Libraries ,03 medical and health sciences ,030104 developmental biology ,Drug Discovery ,Technology integration ,Molecular Medicine ,Profiling (information science) ,Animals ,Humans - Abstract
Small-molecule (SM) leads in the early drug discovery pipeline are progressed primarily based on potency against the intended target(s) and selectivity against a very narrow slice of the proteome. So, why is there a tendency to wait until SMs are matured before probing for a deeper mechanistic understanding? For one, there is a concern about the interpretation of complex -omic data outputs and the resources needed to test these hypotheses. However, with recent advances in broad endpoint profiling assays that have companion reference databases and refined technology integration strategies, we argue that data complexity can translate into meaningful decision-making. This same strategy can also prioritize phenotypic screening hits to increase the likelihood of accessing unprecedented target space. In this Perspective. we will highlight a cohesive process that supports SM hit prosecution, providing a data-driven rationale and a suite of methods for direct identification of SM targets driving relevant biological end points.
- Published
- 2018
17. An activating mutation of the NSD2 histone methyltransferase drives oncogenic reprogramming in acute lymphocytic leukemia
- Author
-
Richard L. Bennett, Qiang Jeremy Wen, Catalina Troche, Alok Swaroop, Alexander D. MacKerell, John D. Crispino, Neil L. Kelleher, Benjamin H. Durham, Marinka Bulic, Christine Will, Wenbo Yu, Xiaoxiao Huang, Jonathan D. Licht, and Jon A. Oyer
- Subjects
0301 basic medicine ,Cancer Research ,Mutant ,Mutation, Missense ,histone ,Article ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,Nucleosome ,Missense mutation ,Humans ,Neoplasm Invasiveness ,Molecular Biology ,relapse ,biology ,epigenetics ,Wild type ,leukemia ,Methylation ,Histone-Lysine N-Methyltransferase ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Cellular Reprogramming ,Cell biology ,Neoplasm Proteins ,Repressor Proteins ,030104 developmental biology ,Histone ,Amino Acid Substitution ,030220 oncology & carcinogenesis ,Histone methyltransferase ,biology.protein ,Heterografts ,methylation ,Reprogramming ,Neoplasm Transplantation ,HeLa Cells - Abstract
NSD2, a histone methyltransferase specific for methylation of histone 3 lysine 36 (H3K36), exhibits a glutamic acid to lysine mutation at residue 1099 (E1099K) in childhood acute lymphocytic leukemia (ALL), and cells harboring this mutation can become the predominant clone in relapsing disease. We studied the effects of this mutant enzyme in silico, in vitro, and in vivo using gene edited cell lines. The E1099K mutation altered enzyme/substrate binding and enhanced the rate of H3K36 methylation. As a result, cell lines harboring E1099K exhibit increased H3K36 dimethylation and reduced H3K27 trimethylation, particularly on nucleosomes containing histone H3.1. Mutant NSD2 cells exhibit reduced apoptosis and enhanced proliferation, clonogenicity, adhesion, and migration. In mouse xenografts, mutant NSD2 cells are more lethal and brain invasive than wildtype cells. Transcriptional profiling demonstrates that mutant NSD2 aberrantly activates factors commonly associated with neural and stromal lineages in addition to signaling and adhesion genes. Identification of these pathways provides new avenues for therapeutic interventions in NSD2 dysregulated malignancies.
- Published
- 2017
18. A Gain of Function Mutation in the NSD2 Histone Methyltransferase Drives Glucocorticoid Resistance Via Blocking Receptor Auto-Induction and BIM/Bmf Expression in ALL
- Author
-
Sharon Norton, Matthew D. Hall, Richard B. Lock, Amin Sobn, Jon A. Oyer, Duohui Jing, Marta Kulis, Jianping Li, Julia Hlavka Zhang, Jonathan H. Shrimp, Richard L. Bennett, Crissandra Pipe, Alok Swaroop, Jonathan D. Licht, Christine Will, Min Shen, Catalina Troche, Alberto Riva, Jacob S. Roth, and Daphné Dupéré-Richer
- Subjects
Regulation of gene expression ,Mutation ,DNA repair ,Cell growth ,Immunology ,Mutant ,Cell Biology ,Hematology ,Biology ,Cell cycle ,medicine.disease_cause ,Biochemistry ,Chromatin ,Cell biology ,Cell culture ,medicine - Abstract
Despite improvements in chemotherapy that have increased the 5-year survival rates of pediatric ALL to close to 90%, 15-20% of patients may relapse with a very poor prognosis. Pediatric ALL patients, particularly those in relapse can harbor a specific point mutation (E1099K) in NSD2 (nuclear receptor binding SET domain protein 2) gene, also known as MMSET or WHSC1, which encodes a histone methyl transferase specific for H3K36me2. To understand the biology of mutant NSD2, we used CRISPR-Cas9 gene editing to disrupt the NSD2E1099K mutant allele in B-ALL cell lines (RCH-ACV and SEM) and T-ALL cell line (RPMI-8402) or insert the E1099K mutation into the NSD2WT T-ALL cell line (CEM) and B-ALL cell line (697). Cell lines in which the NSD2E1099K mutant allele is present display increased global levels of H3K36me2 and decreased H3K27me3. NSD2E1099Kcells demonstrate enhanced cell growth, colony formation and migration. NSD2E1099K mutant cell lines assayed by RNA-Seq exhibit an aberrant gene signature, mostly representing gene activation, with activation of signaling pathways, genes implicated in the epithelial mesenchymal transition and prominent expression of neural genes not generally found in hematopoietic tissues. Accordingly, NSD2E1099K cell lines showed prominent tropism to the central neural system in xenografts. To understand why this NSD2 mutations are identified prominently in children who relapse early from therapy for ALL, we performed high-throughput screening in our isogenic cell lines with the National Center for Advancing Translation Science (NCATS) Pharmaceutical Collection and other annotated chemical libraries and found that NSD2E1099K cells are resistant to glucocorticoids (GC) but not to other chemotherapeutic agents used to treat ALL such as vincristine, doxorubicin, cyclophosphamide, methotrexate, and 6-mercaptopurine. Accordingly, patient-derived-xenograft ALL cells with NSD2E1099K mutation were resistant to GC treatment. Reversion of NSD2E1099K mutation to NSD2WT restored GC sensitivity to both B- and T-ALL cell lines, which was accompanied by cell cycle arrest in G1 and induced-apoptosis. Furthermore, knock-in of the NSD2E1099K mutation conferred GC resistance to ALL cell lines by triggering cell cycle progression, proliferation and anti-apoptotic processes. Mice with NSD2E1099K xenografts were completely resistant to GC treatment while treatment of mice injected with isogenic NSD2WT cells led to significant tumor reduction and survival benefit. To illustrate these biological phenotypes and understand the molecular mechanism of GC resistance driven by NSD2E1099Kmutation, we investigated the GC-induced transcriptome, GC receptor (GR) binding sites and related epigenetic changes in isogenic ALL cell lines in response to GC treatment. RNA-Seq showed that GC transcriptional response was almost completely blocked in NSD2E1099K cells, especially in T-ALL cell lines, correlating with their lack of biological response. GC treatment activated apoptotic pathways and downregulated cell cycle and DNA repair pathways only in NSD2WT cells. The critical pro-apoptotic regulators BIM and BMF failed to be activated by GC in NSD2E1099K cells but were prominently activated when the NSD2 mutation was removed. Chromatin immunoprecipitation sequencing (ChIP-Seq) showed that, the NSD2E1099K mutation blocked the ability of GR and CTCF to bind most GC response elements (GREs) such as those within BIM and BMF. While GR binding in NSD2WT cells was accompanied by increased H3K27 acetylation and gene expression, this failed to occur in NSD2 mutant cells. Furthermore, we found that GR RNA and protein levels were repressed in ALL cells expressing NSD2E1099K and GC failed to induce GR expression in these cells. Paradoxically, while H3K27me3 levels were generally decreased in NSD2E1099K cells, we saw increased levels of H3K27me3 at the GRE within the GR gene body where GR itself and CTCF normally bind, suggesting a novel role for the polycomb repressive complex 2 and EZH2 inhibitors for this form of GC resistance. In conclusion, these studies demonstrate that NSD2E1099K mutation may play an important role in treatment failure of pediatric ALL relapse by interfering with the GR expression and its ability to bind and activate key target genes. Gene editing screens are being performed to understand how to overcome this resistance. Disclosures No relevant conflicts of interest to declare.
- Published
- 2019
- Full Text
- View/download PDF
19. Point mutation E1099K in MMSET/NSD2 enhances its methyltranferase activity and leads to altered global chromatin methylation in lymphoid malignancies
- Author
-
Teresa Ezponda, Jonathan D. Licht, Ari Melnick, Adolfo A. Ferrando, Ross L. Levine, Neil L. Kelleher, Alexander D. MacKerell, Maddalena Allegretta, Yupeng Zheng, Jon A. Oyer, C. I. Okot-Kotber, J. Shim, Zachary Carpenter, Jay P. Patel, Relja Popovic, and Xiaoxiao Huang
- Subjects
Cancer Research ,Lymphoma ,Point mutation ,Repressor ,Histone-Lysine N-Methyltransferase ,Hematology ,Methylation ,DNA Methylation ,Biology ,medicine.disease ,Article ,Chromatin ,Repressor Proteins ,Oncology ,Histone methyltransferase ,DNA methylation ,medicine ,Cancer research ,Humans ,Point Mutation - Abstract
Point mutation E1099K in MMSET/NSD2 enhances its methyltranferase activity and leads to altered global chromatin methylation in lymphoid malignancies
- Published
- 2013
- Full Text
- View/download PDF
20. Aberrantly silenced promoters retain a persistent memory of the silenced state after long-term reactivation
- Author
-
Jon A. Oyer, Mitchell S. Turker, Sarah Godsey, and Phillip A. Yates
- Subjects
Chromatin Immunoprecipitation ,Time Factors ,Health, Toxicology and Mutagenesis ,Adenine Phosphoribosyltransferase ,Decitabine ,Hydroxamic Acids ,Methylation ,Article ,Histones ,Mice ,Cell Line, Tumor ,Genetics ,Animals ,Gene silencing ,Promoter Regions, Genetic ,DNA Modification Methylases ,Molecular Biology ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Lysine ,Promoter ,DNA Methylation ,Molecular biology ,Chromatin ,Gene Expression Regulation, Neoplastic ,Histone Deacetylase Inhibitors ,Histone ,CpG site ,DNA methylation ,Azacitidine ,biology.protein ,CpG Islands ,Chromatin immunoprecipitation - Abstract
A hallmark of aberrant DNA methylation-associated silencing is reversibility. However, long-term stability of reactivated promoters has not been explored. To examine this issue, spontaneous reactivant clones were isolated from mouse embryonal carcinoma cells bearing aberrantly silenced Aprt alleles and re-silencing frequencies were determined as long as three months after reactivation occurred. Despite continuous selection for expression of the reactivated Aprt alleles, exceptionally high spontaneous re-silencing frequencies were observed. A DNA methylation analysis demonstrated retention of sporadic methylation of CpG sites in a protected region of the Aprt promoter in many reactivant alleles suggesting a role for these methylated sites in the re-silencing process. In contrast, a chromatin immunoprecipitation (ChIP) analysis for methyl-H3K4, acetyl-H3K9, and dimethyl-H3K9 levels failed to reveal a specific histone modification that could explain high frequency re-silencing. These results demonstrate that aberrantly silenced and reactivated promoters retain a persistent memory of having undergone the silencing process and suggest the failure to eliminate all CpG methylation as a potential contributing mechanism.
- Published
- 2011
- Full Text
- View/download PDF
21. A Gain of Function Mutation in the NSD2 Histone Methyltransferase Drives Glucocorticoid Resistance of Acute Lymphoblastic Leukemia
- Author
-
Marta Kulis, Jianping Li, Jonathan D. Licht, Alberto Riva, Alok Swaroop, Richard L. Bennett, Crissandra Pipe, Jon A. Oyer, Sharon Norton, Catalina Troche, Daphné Dupéré-Richer, and Christine Will
- Subjects
0301 basic medicine ,Mutation ,Cell growth ,T cell ,Immunology ,Mutant ,Wild type ,Cell Biology ,Hematology ,Cell cycle ,Biology ,medicine.disease_cause ,Biochemistry ,Pediatric cancer ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,medicine - Abstract
Acute lymphoblastic leukemia (ALL) is the most common diagnosed pediatric cancer. Despite improvements in chemotherapy that have increased the 5-year survival rate to close to 90%, 15-20% of these patients may relapse with the majority of such children succumbing to this disease. Pediatric ALL patients, particularly those in relapse can harbor a specific point mutation (E1099K) in NSD2 (nuclear receptor binding SET domain protein 2) gene, also known as MMSET or WHSC1, which encodes a histone methyl transferase specific for H3K36me2. To understand the biological processes mediated by mutant NSD2, we used CRISPR-Cas9 gene editing to disrupt the NSD2E1099K mutant allele in two B-ALL cell lines (RCH-ACV and SEM) and one T-ALL cell line (RPMI-8402) and inserted the E1099K mutation into three ALL cell lines (697, CEM, MOLT4). Cell lines in which the NSD2E1099K mutant allele is present display increased global levels of H3K36me2 and decreased H3K27me3. NSD2E1099Kcells compared to cells in which the mutation is removed demonstrate enhanced cell growth, colony formation and migration. NSD2 mutant cell lines assayed by RNA-Seq exhibit an aberrant gene signature, mostly representing gene activation, with activation of signaling pathways, genes implicated in the epithelial mesenchymal transition and prominent expression of neural genes not generally found in hematopoietic tissues. Accordingly, NSD2E1099K cell lines showed prominent tropism to the central neural system (CNS) in xenografts. The NSD2 mutation is found prominently in children who relapse early from therapy for ALL, and NSD2E1099K cells are particularly resistant to glucocorticoids (GC). Reversion of NSD2E1099K mutation to wild type NSD2 conferred glucocorticoid sensitivity to both B and T cell lines. GC response upon disruption of mutant NSD2 was accompanied by cell cycle arrest and apoptosis. Mice xenografted with NSD2E1099K cells were completely resistant to GC treatment while treatment of mice injected with isogenic NSD2 wild-type cells led to significant tumor reduction and survival extension. RNA-Seq analysis showed that GC transcriptional response was almost completely blocked in NSD2E1099K cells, correlating with their lack of biological response. GC treatment activated apoptotic pathways and downregulated cell cycle and DNA repair pathways only in NSD2 wild-type cells. Furthermore, in NSD2 mutant cells, there was lower basal expression level of glucocorticoid receptor (GR) and GR levels were not significantly induced by GC. Accordingly, after treatment with GC, there was significantly less DNA-binding activity of the GR in NSD2E1099K cells than that of NSD2 wild-type cells. The key pro-apoptotic regulators Bim and BMF failed to be activated by GC in NSD2E1099K cells but were prominently activated when the NSD2 mutation was removed. In conclusion, these studies demonstrate that the NSD2E1099K mutation may play an important role in treatment failure of pediatric ALL relapse by causing GC resistance. Future studies will determine how NSD2 which generally activates genes paradoxically blocks the ability of GC and the GR to induce critical pro-death genes. Disclosures Licht: Celgene: Research Funding.
- Published
- 2018
- Full Text
- View/download PDF
22. Author response: Polycomb- and REST-associated histone deacetylases are independent pathways toward a mature neuronal phenotype
- Author
-
Gail Mandel, Huilan Yao, James C. McGann, Saurabh K. Garg, Lujian Liao, Xin Feng, Jun Liu, John R. Yates, and Jon A Oyer
- Subjects
Histone ,biology ,Neuronal phenotype ,biology.protein ,Rest (music) ,Cell biology - Published
- 2014
- Full Text
- View/download PDF
23. Polycomb- and REST-associated histone deacetylases are independent pathways toward a mature neuronal phenotype
- Author
-
Saurabh K. Garg, James C. McGann, Huilan Yao, Xin Feng, Gail Mandel, Jun Liu, Lujian Liao, John R. Yates, and Jon A Oyer
- Subjects
QH301-705.5 ,Cellular differentiation ,Science ,Repressor ,RE1-silencing transcription factor ,Methylation ,Histone Deacetylases ,General Biochemistry, Genetics and Molecular Biology ,neuronal ,Histones ,Mice ,poised ,Animals ,ES cell ,Biology (General) ,Promoter Regions, Genetic ,Embryonic Stem Cells ,mouse ,Neurons ,Genetics ,General Immunology and Microbiology ,biology ,Lysine ,General Neuroscience ,REST ,Polycomb Repressive Complex 2 ,Cell Differentiation ,General Medicine ,ES cells ,Chromatin ,Repressor Proteins ,Polycomb ,Phenotype ,Developmental Biology and Stem Cells ,Gene Expression Regulation ,Genes and Chromosomes ,histone deacetylase ,biology.protein ,H3K4me3 ,Medicine ,Histone deacetylase activity ,Histone deacetylase ,PRC2 ,Research Article - Abstract
The bivalent hypothesis posits that genes encoding developmental regulators required for early lineage decisions are poised in stem/progenitor cells by the balance between a repressor histone modification (H3K27me3), mediated by the Polycomb Repressor Complex 2 (PRC2), and an activator modification (H3K4me3). In this study, we test whether this mechanism applies equally to genes that are not required until terminal differentiation. We focus on the RE1 Silencing Transcription Factor (REST) because it is expressed highly in stem cells and is an established global repressor of terminal neuronal genes. Elucidation of the REST complex, and comparison of chromatin marks and gene expression levels in control and REST-deficient stem cells, shows that REST target genes are poised by a mechanism independent of Polycomb, even at promoters which bear the H3K27me3 mark. Specifically, genes under REST control are actively repressed in stem cells by a balance of the H3K4me3 mark and a repressor complex that relies on histone deacetylase activity. Thus, chromatin distinctions between pro-neural and terminal neuronal genes are established at the embryonic stem cell stage by two parallel, but distinct, repressor pathways. DOI: http://dx.doi.org/10.7554/eLife.04235.001, eLife digest When an embryo is developing, genes are switched on or off at different times, for many different reasons. Many of these genes are switched off, or repressed, by making the DNA inaccessible to the various proteins and molecules that control gene activity. This is achieved by altering the way that the DNA is packaged into a compacted structure called chromatin. A host of proteins modify the structure of chromatin: it can be made more tightly packaged, which keeps genes switched off; or it can be made more loosely packaged, which allows the genes within to be accessed and switched on. The stem cells in an embryo are able to give rise to many different types of specialized cell. Genes that determine which cell type a stem cell will eventually become are often kept in a so-called ‘poised’ state, and have chromatin modifications that encourage genes to be switch on, as well as modifications that switch genes off. Current thinking is that this poised state allows these genes to be switched on or off rapidly in response to the signals that the cell receives during development. The only known protein complex that causes the chromatin to become more compacted in this poised state is the Polycomb complex. This complex binds to specific regions of DNA and is thought to allow stem cells to remain able to become different cell types by repressing the genes required for adopting a specialized cell fate. However, it is unclear if this poised state also regulates those genes that control the final stages of a cell becoming a specific cell type. McGann et al. investigated genes that are involved in the final stages of a nerve cell's development. These genes are regulated by another protein called REST, which acts to repress the genes in non-neuronal cells. McGann et al. found that the genes that are regulated by REST in embryonic stem cells from mice also have their chromatin modified in two contrasting ways. Some of the modifications are linked to switching genes on, while others are linked to keeping genes switched off. Thus these genes are also in a poised state. However, for these genes, this state is acquired without the activity of the Polycomb complex. The results of McGann et al. show that two similar, but distinct, mechanisms keep the genes required for the early and the late stages of nerve cell development in a poised state. If this poised state aids the development of other cell types (for example muscle or fat cells), uncovering how it is achieved could improve our ability to direct stem cells to develop into specific cell types and tissues. DOI: http://dx.doi.org/10.7554/eLife.04235.002
- Published
- 2014
24. Cardiovascular and Systemic Microvascular Effects of Anti-Vascular Endothelial Growth Factor Therapy for Cancer
- Author
-
Jon A. Oyer, Terry K. Morgan, Stuart Bunting, Jonathan R. Lindner, Aris Xie, Susan P. Bagby, Beat A. Kaufmann, Yue Qi, J. Todd Belcik, Ganesh Kolumam, Sherry Bullens, and Joe Kowalski
- Subjects
Ramipril ,Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,Hemodynamics ,030204 cardiovascular system & hematology ,Kidney ,Article ,Renal Circulation ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Afterload ,Ventricular hypertrophy ,Internal medicine ,Neoplasms ,Medicine ,Animals ,Ventricular remodeling ,Aorta ,030304 developmental biology ,0303 health sciences ,business.industry ,Microcirculation ,Antibodies, Monoclonal ,Stroke volume ,medicine.disease ,contrast echocardiography ,VEGF ,Angiotensin II ,3. Good health ,Mice, Inbred C57BL ,Endocrinology ,Echocardiography ,Pathophysiology of hypertension ,Hypertension ,ventricular hypertrophy ,Cardiology ,Cardiology and Cardiovascular Medicine ,business ,medicine.drug - Abstract
Objectives This study sought to evaluate the contribution of microvascular functional rarefaction and changes in vascular mechanical properties to the development of hypertension and secondary ventricular remodeling that occurs with anti-vascular endothelial growth factor (VEGF) therapy. Background Hypertension is a common side effect of VEGF inhibitors used in cancer medicine. Methods Mice were treated for 5 weeks with an anti-murine VEGF-A monoclonal antibody, antibody plus ramipril, or sham treatment. Microvascular blood flow (MBF) and blood volume (MBV) were quantified by contrast-enhanced ultrasound in skeletal muscle, left ventricle (LV), and kidney. Echocardiography and invasive hemodynamics were used to assess ventricular function, dimensions and vascular mechanical properties. Results Ambulatory blood pressure increased gradually over the first 3 weeks of anti-VEGF therapy. Compared with controls, anti-VEGF–treated mice had similar aortic elastic modulus and histological appearance, but a marked increase in arterial elastance, indicating increased afterload, and elevated plasma angiotensin II. Increased afterload in treated mice led to concentric LV remodeling and reduced stroke volume without impaired LV contractility determined by LV peak change in pressure over time (dp/dt) and the end-systolic dimension–pressure relation. Anti-VEGF therapy did not alter MBF or MBV in skeletal muscle, myocardium, or kidney; but did produce cortical mesangial glomerulosclerosis. Ramipril therapy almost entirely prevented the adverse hemodynamic effects, increased afterload, and LV remodeling in anti-VEGF–treated mice. Conclusions Neither reduced functional microvascular density nor major alterations in arterial mechanical properties are primary causes of hypertension during anti-VEGF therapy. Inhibition of VEGF leads to an afterload mismatch state, increased angiotensin II, and LV remodeling, which are all ameliorated by angiotensin-converting enzyme inhibition.
- Published
- 2012
25. Allele-Specific Crispr Targeting Reveals Epigenetic and Phenotypic Effects of a MMSET Gain of Function Mutation Found in Relapsed Acute Lymphoblastic Leukemia
- Author
-
Teresa Ezponda, Neil L. Kelleher, Jonathan D. Licht, Xiaoxiao Huang, Jon A. Oyer, Christine Will, Relja Popovic, and Yupeng Zheng
- Subjects
Mutation ,Methyltransferase ,Immunology ,EZH2 ,Mutant ,Wild type ,Cell Biology ,Hematology ,Biology ,medicine.disease_cause ,Biochemistry ,Pediatric cancer ,medicine ,Cancer research ,Epigenetics ,Allele - Abstract
Chromatin regulators including methyltransferases are frequently disrupted by inactivating mutations in a wide spectrum of cancers. In contrast gain of function mutations that increase enzyme activity are much more rare and found in only two enzymes within the family of approximately 60 SET domain methyltransferases, MMSET and EZH2. Specific recurrent mutations in either MMSET or EZH2 are detected in hematological malignancies in conjunction with increased methylation of their histone substrates, H3K36 and H3K27, respectively. We and others characterized the MMSET E1099K mutant, which generates a clear molecular hallmark of increased dimethylation of H3K36 (H3K36me2). This gain of function also induces a corresponding decrease in trimethylation at H3K27 (H3K27me3). The altered balance between H3K36me2 and H3K27me3 mimics global chromatin effects that we previously reported in multiple myeloma lines that overexpress wild type MMSET due to the t(4:14) translocation. Research led by the Pediatric Cancer Genome Project has shown that MMSET E1099K is detected in ~10% of B-cell acute lymphoblastic leukemia (ALL) and particularly enriched in samples following disease relapse. Therefore, we hypothesize that this mutation is a driver of ALL progression and mediates resistance to therapy. To address this question, we have rescued or disrupted the endogenous E1099K mutation in cell lines from relapsed ALL and characterized the resulting molecular and phenotypic effects. The sequence context of the E1099K mutation allows allele-specific CRISPR targeting, which we have exploited to create isogenic lines that differ by MMSET E1099K status. We isolated subclones from three parental lines and sequencing analysis revealed distinct outcomes: indel mutations that disrupt the E1099K allele or rescue to WT by interchromosomal gene conversion. Comparing these subclones to non-targeted controls showed that loss of E1099K causes a 3-fold reduction in H3K36me2 levels and 5-fold increase in H3K27me3. Mass spectrometry-based measurement of methylation kinetics revealed that E1099K accelerates the rate constant of conversion from unmodified H3K36 to monomethylation. This corresponds to an increased ability of the mutant enzyme to turnover substrate in vitro. Because H3K36 and H3K27 methylation both contribute to transcriptional regulation, we compared expression profiles and identified a common set of genes overexpressed in cells lines harboring the mutant allele. These overexpressed genes included components of the SLIT/ROBO, WNT/Beta-Catenin, and cell adhesion pathways. Correlating with these expression changes, several phenotypic changes resulted from loss of E1099K, such as reduction in proliferation, colony-forming ability, and adhesive properties. Loss of E1099K also increased sensitivity to chemotherapeutic agents used to treat ALL, such as doxorubicin and dexamethasone. One gene consistently upregulated in the presence of E1099K was the transcription factor ETV1. In support of ETV1 being a key mediator of E1099K-driven phenotypes we found that parental lines treated with an ETV1 inhibitor displayed reduced viability and adhesion. To further our phenotypic analysis, subclones and parental lines have been tagged with luciferase or fluorescent markers to assess invasion in models of metastasis and allow in vivo monitoring of tumors. Collectively, we have developed gene-targeting reagents specific for the MMSET E1099K mutation and used these tools to show its impact on global chromatin environment and cell phenotypes. Disclosures No relevant conflicts of interest to declare.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.