44 results on '"Dylan M. Marchione"'
Search Results
2. H3K27M in Gliomas Causes a One-Step Decrease in H3K27 Methylation and Reduced Spreading within the Constraints of H3K36 Methylation
- Author
-
Ashot S. Harutyunyan, Haifen Chen, Tianyuan Lu, Cynthia Horth, Hamid Nikbakht, Brian Krug, Caterina Russo, Eric Bareke, Dylan M. Marchione, Mariel Coradin, Benjamin A. Garcia, Nada Jabado, and Jacek Majewski
- Subjects
pediatric high-grade glioma ,H3.3K27M ,epigenomics ,histone methylation ,computational modeling ,Biology (General) ,QH301-705.5 - Abstract
Summary: The discovery of H3K27M mutations in pediatric gliomas marked a new chapter in cancer epigenomics. Numerous studies have investigated the effect of this mutation on H3K27 trimethylation, but only recently have we started to realize its additional effects on the epigenome. Here, we use isogenic glioma H3K27M+/− cell lines to investigate H3K27 methylation and its interaction with H3K36 and H3K9 modifications. We describe a “step down” effect of H3K27M on the distribution of H3K27 methylation: me3 is reduced to me2, me2 is reduced to me1, whereas H3K36me2/3 delineates the boundaries for the spread of H3K27me marks. We also observe a replacement of H3K27me2/3 silencing by H3K9me3. Using a computational simulation, we explain our observations by reduced effectiveness of PRC2 and constraints imposed on the deposition of H3K27me by antagonistic H3K36 modifications. Our work further elucidates the effects of H3K27M in gliomas as well as the general principles of deposition in H3K27 methylation.
- Published
- 2020
- Full Text
- View/download PDF
3. Characterization of histone acylations links chromatin modifications with metabolism
- Author
-
Johayra Simithy, Simone Sidoli, Zuo-Fei Yuan, Mariel Coradin, Natarajan V. Bhanu, Dylan M. Marchione, Brianna J. Klein, Gleb A. Bazilevsky, Cheryl E. McCullough, Robert S. Magin, Tatiana G. Kutateladze, Nathaniel W. Snyder, Ronen Marmorstein, and Benjamin A. Garcia
- Subjects
Science - Abstract
A number of histone lysine modifications related to acetylation have been identified, but their functional significance is unclear. Here, the authors use in vitro and in vivo assays to characterize eight acyl histone post-translational modifications and link their abundance with metabolism.
- Published
- 2017
- Full Text
- View/download PDF
4. P441: CLINICAL AND MOLECULAR CHARACTERISTICS OF AML PATIENTS WITH AN EXCEPTIONAL RESPONSE TO IVOSIDENIB
- Author
-
Justin M. Watts, Prapti A Patel, Sung Choe, Xiaofei Bai, Dylan M Marchione, and Eytan Stein
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
5. P724: UPDATED SUBSTUDY RESULTS FOR IVOSIDENIB IN IDH1-MUTANT RELAPSED/REFRACTORY MYELODYSPLASTIC SYNDROME
- Author
-
Courtney Dinardo, Gail Roboz, Justin M. Watts, Yazan Madanat, Gabrielle T Prince, Praneeth Baratam, Stéphane de Botton, Anthony Stein, James M Foran, Martha L Arellano, David Sallman, Dylan M Marchione, Xiaofei Bai, Prapti A Patel, Stephanie M Kapsalis, Guillermo Garcia-Manero, and Amir Fathi
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
6. Multicenter Phase I Trial of Ivosidenib as Maintenance Treatment Following Allogeneic Hematopoietic Cell Transplantation for IDH1-Mutated Acute Myeloid Leukemia
- Author
-
Amir T. Fathi, Haesook T. Kim, Robert J. Soiffer, Mark J. Levis, Shuli Li, Annette S. Kim, Zachariah DeFilipp, Areej El-Jawahri, Steve L. McAfee, Andrew M. Brunner, Philip C. Amrein, Alice S. Mims, Laura W. Knight, Devon Kelley, AJ S. Bottoms, Lindsey H. Perry, Jonathan L. Wahl, Jennifer Brock, Elayne Breton, Dylan M. Marchione, Vincent T. Ho, and Yi-Bin Chen
- Subjects
Cancer Research ,Oncology - Abstract
Purpose: Isocitrate dehydrogenase 1 (IDH1) mutations occur in 5% to 10% of patients with acute myeloid leukemia (AML). Ivosidenib is an IDH1 inhibitor, approved for use in patients with IDH1-mutated AML. Patients and Methods: We conducted a multicenter, phase I trial of maintenance ivosidenib following allogeneic hematopoietic cell transplantation (HCT) in patients with IDH1-mutated AML. Ivosidenib was initiated between days 30 and 90 following HCT and continued for up to 12 28-day cycles. The first dose level was 500 mg daily, with level reduction to 250 mg daily, if needed, in a 3 × 3 de-escalation design. Ten additional patients would then receive the MTD or recommended phase 2 dose (RP2D). The primary endpoint was establishing the MTD or RP2D of ivosidenib. Results: Eighteen patients were enrolled, of whom 16 initiated post-HCT ivosidenib. One dose-limiting toxicity, grade(g) 3 QTc prolongation, was observed. The RP2D was established at 500 mg daily. Attributable g≥3 adverse events were uncommon, with the most common being QTc prolongation in 2 patients. Eight patients discontinued maintenance, with only one due to adverse event. Six-month cumulative incidence (CI) of gII-IV aGVHD was 6.3%, and 2-year CI of all cGVHD was 63%. Two-year CI of relapse and nonrelapse mortality (NRM) were 19% and 0%, respectively. Two-year progression-free (PFS) was 81%, and 2-year overall survival (OS) was 88%. Conclusions: Ivosidenib is safe and well-tolerated as maintenance therapy following HCT. Cumulative incidence of relapse and NRM, as well as estimations of PFS and OS, were promising in this phase I study.
- Published
- 2023
7. Molecular Characterization of Clinical Response and Relapse in Patients with IDH1m ND-AML Treated with Ivo+AZA in the AGILE Study
- Author
-
Hartmut Döhner, Dylan M. Marchione, Sung Choe, Pau Montesinos, Christian Recher, Susana Vives, Ewa Zarzycka, Jianxiang Wang, Claudio Cerchione, Michael Heuser, Rodrigo T. Calado, Andre C. Schuh, Su-Peng Yeh, Adolfo De La Fuente, Jianan Hui, Prapti Patel, Diego A. Gianolio, Scott R. Daigle, Courtney D. DiNardo, and Stephane De Botton
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
8. Supplemental Table 1 from Multicenter Phase I Trial of Ivosidenib as Maintenance Treatment Following Allogeneic Hematopoietic Cell Transplantation for IDH1-Mutated Acute Myeloid Leukemia
- Author
-
Yi-Bin Chen, Vincent T. Ho, Dylan M. Marchione, Elayne Breton, Jennifer Brock, Jonathan L. Wahl, Lindsey H. Perry, AJ S. Bottoms, Devon Kelley, Laura W. Knight, Alice S. Mims, Philip C. Amrein, Andrew M. Brunner, Steve L. McAfee, Areej El-Jawahri, Zachariah DeFilipp, Annette S. Kim, Shuli Li, Mark J. Levis, Robert J. Soiffer, Haesook T. Kim, and Amir T. Fathi
- Abstract
Representativeness of Study Participants
- Published
- 2023
9. Supplemental Figure 2 from Multicenter Phase I Trial of Ivosidenib as Maintenance Treatment Following Allogeneic Hematopoietic Cell Transplantation for IDH1-Mutated Acute Myeloid Leukemia
- Author
-
Yi-Bin Chen, Vincent T. Ho, Dylan M. Marchione, Elayne Breton, Jennifer Brock, Jonathan L. Wahl, Lindsey H. Perry, AJ S. Bottoms, Devon Kelley, Laura W. Knight, Alice S. Mims, Philip C. Amrein, Andrew M. Brunner, Steve L. McAfee, Areej El-Jawahri, Zachariah DeFilipp, Annette S. Kim, Shuli Li, Mark J. Levis, Robert J. Soiffer, Haesook T. Kim, and Amir T. Fathi
- Abstract
Supplemental Figure 2: Overall and progression-free survival for patients according to NPM1 mutational status
- Published
- 2023
10. Data from Multicenter Phase I Trial of Ivosidenib as Maintenance Treatment Following Allogeneic Hematopoietic Cell Transplantation for IDH1-Mutated Acute Myeloid Leukemia
- Author
-
Yi-Bin Chen, Vincent T. Ho, Dylan M. Marchione, Elayne Breton, Jennifer Brock, Jonathan L. Wahl, Lindsey H. Perry, AJ S. Bottoms, Devon Kelley, Laura W. Knight, Alice S. Mims, Philip C. Amrein, Andrew M. Brunner, Steve L. McAfee, Areej El-Jawahri, Zachariah DeFilipp, Annette S. Kim, Shuli Li, Mark J. Levis, Robert J. Soiffer, Haesook T. Kim, and Amir T. Fathi
- Abstract
Purpose:Isocitrate dehydrogenase 1 (IDH1) mutations occur in 5% to 10% of patients with acute myeloid leukemia (AML). Ivosidenib is an IDH1 inhibitor, approved for use in patients with IDH1-mutated AML.Methods:We conducted a multicenter, phase I trial of maintenance ivosidenib following allogeneic hematopoietic cell transplantation (HCT) in patients with IDH1-mutated AML. Ivosidenib was initiated between days 30 and 90 following HCT and continued for up to 12 28-day cycles. The first dose level was 500 mg daily, with level reduction to 250 mg daily, if needed, in a 3 × 3 de-escalation design. Ten additional patients would then receive the MTD or recommended phase 2 dose (RP2D). The primary endpoint was establishing the MTD or RP2D of ivosidenib.Results:Eighteen patients were enrolled, of whom 16 initiated post-HCT ivosidenib. One dose-limiting toxicity, grade(g) 3 QTc prolongation, was observed. The RP2D was established at 500 mg daily. Attributable g≥3 adverse events were uncommon, with the most common being QTc prolongation in 2 patients. Eight patients discontinued maintenance, with only one due to adverse event. Six-month cumulative incidence (CI) of gII-IV aGVHD was 6.3%, and 2-year CI of all cGVHD was 63%. Two-year CI of relapse and nonrelapse mortality (NRM) were 19% and 0%, respectively. Two-year progression-free (PFS) was 81%, and 2-year overall survival (OS) was 88%.Conclusions:Ivosidenib is safe and well-tolerated as maintenance therapy following HCT. Cumulative incidence of relapse and NRM, as well as estimations of PFS and OS, were promising in this phase I study.
- Published
- 2023
11. Supplemental Figure 1 from Multicenter Phase I Trial of Ivosidenib as Maintenance Treatment Following Allogeneic Hematopoietic Cell Transplantation for IDH1-Mutated Acute Myeloid Leukemia
- Author
-
Yi-Bin Chen, Vincent T. Ho, Dylan M. Marchione, Elayne Breton, Jennifer Brock, Jonathan L. Wahl, Lindsey H. Perry, AJ S. Bottoms, Devon Kelley, Laura W. Knight, Alice S. Mims, Philip C. Amrein, Andrew M. Brunner, Steve L. McAfee, Areej El-Jawahri, Zachariah DeFilipp, Annette S. Kim, Shuli Li, Mark J. Levis, Robert J. Soiffer, Haesook T. Kim, and Amir T. Fathi
- Abstract
Supplemental Figure 1: Overall and progression-free survival for patients, according to whether the transplant followed first line or second line treatment.
- Published
- 2023
12. Table S2 from H3.3 G34W Promotes Growth and Impedes Differentiation of Osteoblast-Like Mesenchymal Progenitors in Giant Cell Tumor of Bone
- Author
-
Nada Jabado, Claudia L. Kleinman, Livia Garzia, Michael D. Taylor, Stephen C. Mack, Benjamin A. Garcia, Peter W. Lewis, Pierre Thibault, Jay S. Wunder, Robert Turcotte, Brendan C. Dickson, Jason Karamchandani, Sungmi Jung, Ashot S. Harutyunyan, Véronique Lisi, Robert Eveleigh, Tianna S. Sihota, Kateryna Rossokhata, Siddhant U. Jain, Takeaki Ishii, Éric Bonneil, Joel Lanoix, Dylan M. Marchione, Damien Faury, Leonie G. Mikael, Carol C.L. Chen, Wajih Jawhar, Liam D. Hendrikse, Shriya Deshmukh, Nicolas De Jay, and Sima Khazaei
- Abstract
Differential Expression in Isogenic Lines
- Published
- 2023
13. Data from H3.3 G34W Promotes Growth and Impedes Differentiation of Osteoblast-Like Mesenchymal Progenitors in Giant Cell Tumor of Bone
- Author
-
Nada Jabado, Claudia L. Kleinman, Livia Garzia, Michael D. Taylor, Stephen C. Mack, Benjamin A. Garcia, Peter W. Lewis, Pierre Thibault, Jay S. Wunder, Robert Turcotte, Brendan C. Dickson, Jason Karamchandani, Sungmi Jung, Ashot S. Harutyunyan, Véronique Lisi, Robert Eveleigh, Tianna S. Sihota, Kateryna Rossokhata, Siddhant U. Jain, Takeaki Ishii, Éric Bonneil, Joel Lanoix, Dylan M. Marchione, Damien Faury, Leonie G. Mikael, Carol C.L. Chen, Wajih Jawhar, Liam D. Hendrikse, Shriya Deshmukh, Nicolas De Jay, and Sima Khazaei
- Abstract
Glycine 34-to-tryptophan (G34W) substitutions in H3.3 arise in approximately 90% of giant cell tumor of bone (GCT). Here, we show H3.3 G34W is necessary for tumor formation. By profiling the epigenome, transcriptome, and secreted proteome of patient samples and tumor-derived cells CRISPR–Cas9-edited for H3.3 G34W, we show that H3.3K36me3 loss on mutant H3.3 alters the deposition of the repressive H3K27me3 mark from intergenic to genic regions, beyond areas of H3.3 deposition. This promotes redistribution of other chromatin marks and aberrant transcription, altering cell fate in mesenchymal progenitors and hindering differentiation. Single-cell transcriptomics reveals that H3.3 G34W stromal cells recapitulate a neoplastic trajectory from a SPP1+ osteoblast-like progenitor population toward an ACTA2+ myofibroblast-like population, which secretes extracellular matrix ligands predicted to recruit and activate osteoclasts. Our findings suggest that H3.3 G34W leads to GCT by sustaining a transformed state in osteoblast-like progenitors, which promotes neoplastic growth, pathologic recruitment of giant osteoclasts, and bone destruction.Significance:This study shows that H3.3 G34W drives GCT tumorigenesis through aberrant epigenetic remodeling, altering differentiation trajectories in mesenchymal progenitors. H3.3 G34W promotes in neoplastic stromal cells an osteoblast-like progenitor state that enables undue interactions with the tumor microenvironment, driving GCT pathogenesis. These epigenetic changes may be amenable to therapeutic targeting in GCT.See related commentary by Licht, p. 1794.This article is highlighted in the In This Issue feature, p. 1775
- Published
- 2023
14. Supplementary Figures from H3.3 G34W Promotes Growth and Impedes Differentiation of Osteoblast-Like Mesenchymal Progenitors in Giant Cell Tumor of Bone
- Author
-
Nada Jabado, Claudia L. Kleinman, Livia Garzia, Michael D. Taylor, Stephen C. Mack, Benjamin A. Garcia, Peter W. Lewis, Pierre Thibault, Jay S. Wunder, Robert Turcotte, Brendan C. Dickson, Jason Karamchandani, Sungmi Jung, Ashot S. Harutyunyan, Véronique Lisi, Robert Eveleigh, Tianna S. Sihota, Kateryna Rossokhata, Siddhant U. Jain, Takeaki Ishii, Éric Bonneil, Joel Lanoix, Dylan M. Marchione, Damien Faury, Leonie G. Mikael, Carol C.L. Chen, Wajih Jawhar, Liam D. Hendrikse, Shriya Deshmukh, Nicolas De Jay, and Sima Khazaei
- Abstract
Supplementary Figures
- Published
- 2023
15. Supplementary Data from H3.3 G34W Promotes Growth and Impedes Differentiation of Osteoblast-Like Mesenchymal Progenitors in Giant Cell Tumor of Bone
- Author
-
Nada Jabado, Claudia L. Kleinman, Livia Garzia, Michael D. Taylor, Stephen C. Mack, Benjamin A. Garcia, Peter W. Lewis, Pierre Thibault, Jay S. Wunder, Robert Turcotte, Brendan C. Dickson, Jason Karamchandani, Sungmi Jung, Ashot S. Harutyunyan, Véronique Lisi, Robert Eveleigh, Tianna S. Sihota, Kateryna Rossokhata, Siddhant U. Jain, Takeaki Ishii, Éric Bonneil, Joel Lanoix, Dylan M. Marchione, Damien Faury, Leonie G. Mikael, Carol C.L. Chen, Wajih Jawhar, Liam D. Hendrikse, Shriya Deshmukh, Nicolas De Jay, and Sima Khazaei
- Abstract
Supplementary Information
- Published
- 2023
16. Supplementary Tables S1-S5 from Epigenomic Reordering Induced by Polycomb Loss Drives Oncogenesis but Leads to Therapeutic Vulnerabilities in Malignant Peripheral Nerve Sheath Tumors
- Author
-
Benjamin A. Garcia, Jacek Majewski, Amanda Lisby, Anissa Djedid, Simone Sidoli, Dylan M. Marchione, and John B. Wojcik
- Abstract
Tabbed excel spreadsheet with each tab representing a single supplementary table. The first cell of each tab contains a description of the contents. Supplemental Table 1: Log2 normalized protein abundances of proteins identified in FFPE MPNST samples. Supplemental Table 2: GO term enrichment for proteins showing increased levels with PRC2 loss. Supplemental Table 3: GO term enrichment for proteins showing decreased levels with PRC2 loss. Supplemental Table 4: GO term enrichment for proteins showing increased levels following both SUZ12 restoration and NSD2 knockdown. Supplemental Table 5: GO term enrichment for proteins showing increased levels following decitabine treatment.
- Published
- 2023
17. Supplementary Figures S1, S2, S3, S4 from Epigenomic Reordering Induced by Polycomb Loss Drives Oncogenesis but Leads to Therapeutic Vulnerabilities in Malignant Peripheral Nerve Sheath Tumors
- Author
-
Benjamin A. Garcia, Jacek Majewski, Amanda Lisby, Anissa Djedid, Simone Sidoli, Dylan M. Marchione, and John B. Wojcik
- Abstract
Merged pdf file containing supplementary figures 1-4
- Published
- 2023
18. Supplementary Data Legends from Epigenomic Reordering Induced by Polycomb Loss Drives Oncogenesis but Leads to Therapeutic Vulnerabilities in Malignant Peripheral Nerve Sheath Tumors
- Author
-
Benjamin A. Garcia, Jacek Majewski, Amanda Lisby, Anissa Djedid, Simone Sidoli, Dylan M. Marchione, and John B. Wojcik
- Abstract
Word file containing legends for supplemental figures S1-S4 and Supplemental tables 1-5
- Published
- 2023
19. Data from Epigenomic Reordering Induced by Polycomb Loss Drives Oncogenesis but Leads to Therapeutic Vulnerabilities in Malignant Peripheral Nerve Sheath Tumors
- Author
-
Benjamin A. Garcia, Jacek Majewski, Amanda Lisby, Anissa Djedid, Simone Sidoli, Dylan M. Marchione, and John B. Wojcik
- Abstract
Malignant peripheral nerve sheath tumor (MPNST) is an aggressive sarcoma with recurrent loss-of-function alterations in polycomb-repressive complex 2 (PRC2), a histone-modifying complex involved in transcriptional silencing. To understand the role of PRC2 loss in pathogenesis and identify therapeutic targets, we conducted parallel global epigenomic and proteomic analysis of archival formalin-fixed, paraffin-embedded (FFPE) human MPNST with and without PRC2 loss (MPNSTLOSS vs. MPNSTRET). Loss of PRC2 resulted in increased histone posttranslational modifications (PTM) associated with active transcription, most notably H3K27Ac and H3K36me2, whereas repressive H3K27 di- and trimethylation (H3K27me2/3) marks were globally lost without a compensatory gain in other repressive PTMs. Instead, DNA methylation globally increased in MPNSTLOSS. Epigenomic changes were associated with upregulation of proteins in growth pathways and reduction in IFN signaling and antigen presentation, suggesting a role for epigenomic changes in tumor progression and immune evasion, respectively. These changes also resulted in therapeutic vulnerabilities. Knockdown of NSD2, the methyltransferase responsible for H3K36me2, restored MHC expression and induced interferon pathway expression in a manner similar to PRC2 restoration. MPNSTLOSS were also highly sensitive to DNA methyltransferase and histone deacetylase (HDAC) inhibitors. Overall, these data suggest that global loss of PRC2-mediated repression renders MPNST differentially dependent on DNA methylation to maintain transcriptional integrity and makes them susceptible to therapeutics that promote aberrant transcription initiation.Significance:Global profiling of histone PTMs and protein expression in archival human MPNST illustrates how PRC2 loss promotes oncogenesis but renders tumors vulnerable to pharmacologic modulation of transcription.See related commentary by Natarajan and Venneti, p. 3172
- Published
- 2023
20. Histone H3K27 dimethyl loss is highly specific for malignant peripheral nerve sheath tumor and distinguishes true PRC2 loss from isolated H3K27 trimethyl loss
- Author
-
Ivan Chebib, Ana B. Larque, Amanda Lisby, John Wojcik, Angela N. Viaene, MacLean Nasrallah, Erik A. Williams, Dylan M. Marchione, Benjamin A. Garcia, Li-Ping Wang, and Mariarita Santi
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Malignant peripheral nerve sheath tumor ,macromolecular substances ,Article ,Pathology and Forensic Medicine ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Biomarkers, Tumor ,medicine ,Humans ,biology ,business.industry ,Polycomb Repressive Complex 2 ,DNA Methylation ,medicine.disease ,Staining ,030104 developmental biology ,Histone ,medicine.anatomical_structure ,Neurofibrosarcoma ,Cell culture ,030220 oncology & carcinogenesis ,biology.protein ,Immunohistochemistry ,Differential diagnosis ,PRC2 ,business ,Merkel cell - Abstract
Malignant peripheral nerve sheath tumors contain loss of histone H3K27 trimethylation (H3K27me3) due to driver mutations affecting the polycomb repressive complex 2 (PRC2). Consequently, loss of H3K27me3 staining has served as a diagnostic marker for this tumor type. However, recent reports demonstrate H3K27me3 loss in numerous other tumors, including some in the differential diagnosis of malignant peripheral nerve sheath tumor. Since these tumors lose H3K27me3 through mechanisms distinct from PRC2 loss, we set out to determine whether loss of dimethylation of H3K27, which is also catalyzed by PRC2, might be a more specific marker of PRC2 loss and malignant peripheral nerve sheath tumor. Using mass spectrometry, we identify a near complete loss of H3K27me2 in malignant peripheral nerve sheath tumors and cell lines. Immunohistochemical analysis of 72 malignant peripheral nerve sheath tumors, seven K27M-mutant gliomas, 43 ependymomas, and 10 Merkel cell carcinomas demonstrates that while H3K27me3 loss is common across these tumor types, H3K27me2 loss is limited to malignant peripheral nerve sheath tumors and is highly concordant with H3K27me3 loss (33/34 cases). Thus, increased specificity does not come at the cost of greatly reduced sensitivity. To further compare H3K27me2 and H3K27me3 immunohistochemistry, we investigated 42 melanomas and 54 synovial sarcomas, histologic mimics of malignant peripheral nerve sheath tumor with varying degrees of H3K27me3 loss in prior reports. While global H3K27me3 loss was not seen in these tumors, weak and limited H3K27me3 staining was common. By contrast, H3K27me2 staining was more clearly retained in all cases, making it a superior binary classifier. This was confirmed by digital image analysis of stained slides. Our findings indicate that H3K27me2 loss is highly specific for PRC2 loss and that PRC2 loss is a rarer phenomenon than H3K27me3 loss. Consequently, H3K27me2 loss is a superior diagnostic marker for malignant peripheral nerve sheath tumor.
- Published
- 2019
21. The histone mark H3K36me2 recruits DNMT3A and shapes the intergenic DNA methylation landscape
- Author
-
Marcus A. Cheek, Michael-Christopher Keogh, John T. McGuire, Matthew R. Marunde, Daniel N. Weinberg, Simon Papillon-Cavanagh, Kartik N. Rajagopalan, Chao Lu, Haitao Li, Nada Jabado, Jacek Majewski, Yuan Yue, Anissa Djedid, Eric Bareke, Xinjing Xu, Ashot S. Harutyunyan, Xiao Chen, Agata E. Lemiesz, Cynthia Horth, Hamid Nikbakht, Matthew J. Meiners, Haifen Chen, Dylan M. Marchione, Benjamin A. Garcia, and C. David Allis
- Subjects
0301 basic medicine ,Methyltransferase ,DNA methyltransferase ,Article ,Cell Line ,DNA Methyltransferase 3A ,Histones ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Intergenic region ,Protein Domains ,Animals ,Humans ,DNA (Cytosine-5-)-Methyltransferases ,Growth Disorders ,Genetics ,Multidisciplinary ,Sotos Syndrome ,biology ,Methylation ,DNA Methylation ,Protein Transport ,030104 developmental biology ,Histone ,Histone methyltransferase ,embryonic structures ,DNA methylation ,biology.protein ,DNMT1 ,DNA, Intergenic ,030217 neurology & neurosurgery ,Genome-Wide Association Study ,Protein Binding - Abstract
Enzymes that catalyse CpG methylation in DNA, including the DNA methyltransferases 1 (DNMT1), 3A (DNMT3A) and 3B (DNMT3B), are indispensable for mammalian tissue development and homeostasis1–4. They are also implicated in human developmental disorders and cancers5–8, supporting the critical role of DNA methylation in the specification and maintenance of cell fate. Previous studies have suggested that post-translational modifications of histones are involved in specifying patterns of DNA methyltransferase localization and DNA methylation at promoters and actively transcribed gene bodies9–11. However, the mechanisms that control the establishment and maintenance of intergenic DNA methylation remain poorly understood. Tatton–Brown–Rahman syndrome (TBRS) is a childhood overgrowth disorder that is defined by germline mutations in DNMT3A. TBRS shares clinical features with Sotos syndrome (which is caused by haploinsufficiency of NSD1, a histone methyltransferase that catalyses the dimethylation of histone H3 at K36 (H3K36me2)8,12,13), which suggests that there is a mechanistic link between these two diseases. Here we report that NSD1-mediated H3K36me2 is required for the recruitment of DNMT3A and maintenance of DNA methylation at intergenic regions. Genome-wide analysis shows that the binding and activity of DNMT3A colocalize with H3K36me2 at non-coding regions of euchromatin. Genetic ablation of Nsd1 and its paralogue Nsd2 in mouse cells results in a redistribution of DNMT3A to H3K36me3-modified gene bodies and a reduction in the methylation of intergenic DNA. Blood samples from patients with Sotos syndrome and NSD1-mutant tumours also exhibit hypomethylation of intergenic DNA. The PWWP domain of DNMT3A shows dual recognition of H3K36me2 and H3K36me3 in vitro, with a higher binding affinity towards H3K36me2 that is abrogated by TBRS-derived missense mutations. Together, our study reveals a trans-chromatin regulatory pathway that connects aberrant intergenic CpG methylation to human neoplastic and developmental overgrowth. H3K36me2 targets DNMT3A to intergenic regions and this process, together with H3K36me3-mediated recruitment of DNMT3B, has a key role in establishing and maintaining genomic DNA methylation landscapes.
- Published
- 2019
22. Molecular characterization of clinical response in patients with newly diagnosed acute myeloid leukemia treated with ivosidenib + azacitidine compared to placebo + azacitidine
- Author
-
Stéphane De Botton, Sung Choe, Dylan M. Marchione, Pau Montesinos, Christian Recher, Susana Vives Polo, Ewa Zarzycka, Jianxiang Wang, Giambattista Bertani, Michael Heuser, Rodrigo T. Calado, Andre C. Schuh, Su-Peng Yeh, Jianan Hui, Shuchi Sumant Pandya, Diego A. Gianolio, Scott Daigle, Courtney Denton Dinardo, and Hartmut Dohner
- Subjects
Cancer Research ,Oncology - Abstract
7019 Background: Acute myeloid leukemia (AML) is a disease with a dynamic mutational landscape; 6–10% of patients (pts) have somatic mutations in isocitrate dehydrogenase 1 ( IDH1), which can drive oncogenesis. Ivosidenib (IVO) is a potent oral targeted inhibitor of mutant IDH1 (mIDH1). IVO 500 mg QD + azacitidine (AZA) 75 mg/m2 SC or IV for 7 days in 28-day cycles was shown to significantly improve event-free survival (HR = 0.33 [95% CI 0.16, 0.69], p = 0.0011), median overall survival (24.0 vs 7.9 months), and complete remission + partial hematologic recovery rates (CR/CRh; 52.8% vs 17.6%) vs placebo (PBO) + AZA in the double-blind phase 3 AGILE study (NCT03173248) in pts with newly diagnosed IDH1-mutated AML (ND-AML). IDH1-mutation clearance ( IDH1-MC) and baseline co-mutation analysis from AGILE is reported. Methods: Genomic DNA from bone marrow mononuclear cells (BMMCs) or peripheral blood mononuclear cells (PBMCs), and/or bone marrow aspirate (BMA) were used for molecular studies. IDH1-MC analysis on BMMCs was performed by BEAMing digital PCR (limit of detection 0.02%-0.04%). BMA, BMMCs and PBMCs were utilized for co-mutational analysis by next-generation sequencing, ACE Extended Cancer Panel (detection limit 2%). Results: 146 pts were randomized: 72 to IVO+AZA; 74 to PBO+AZA. Median (range) baseline m IDH1 variant allele frequency in BMMCs was 36.7% (3.1–50.5) in the IVO+AZA arm and 35.5% (3.0–48.6) in the PBO+AZA arm. Updated IDH1-MC data (October 2021) from 47 IVO+AZA and 32 PBO+AZA treated pts with at least 1 on-treatment sample demonstrated IDH1-MC in 21/35 (60%) IVO+AZA pts achieving CR/CRh vs 4/11 (36%) PBO+AZA pts. In CR/CRh pts with time points available after IDH1-MC, suppression of the m IDH1 was durable and IDH1-MC maintained in all subsequent samples in 17/17 (100%) IVO+AZA treated pts and 1/3 (33%) PBO+AZA pts. Further analysis of baseline co-mutations on 120 pts (IVO+AZA: n = 58; PBO+AZA: n = 62) showed that DNMT3A, SRSF2, and RUNX1 were the most frequent in both treatment arms. Importantly, comparison of CR/CRh and non CR/CRh responses by cohort did not identify any single gene or pathway associated with an inferior outcome in IVO+AZA pts compared to PBO+AZA pts (p < 0.05, Fisher’s Exact test). Several genes ( DNMT3A, RUNX1, SRSF2, STAG2) and pathways (Differentiation, Epigenetics, Splicing) were associated with improved outcomes with IVO+AZA, including the RTK pathway, which was previously reported to be associated with primary resistance to IVO monotherapy. Further analysis of patient subgroups, including R132 variants (i.e., R132C vs R132S), will be presented. Conclusions: These data suggest that improved clinical outcomes with IVO+AZA are associated with sustained clearance of the m IDH1 clone including pts with disease that harbor mutations implicated in resistance to IVO monotherapy (e.g., with RTK pathway mutations). Clinical trial information: NCT03173248.
- Published
- 2022
23. Re: Human Chimeric Antigen Receptor Macrophages for Cancer Immunotherapy
- Author
-
Benjamin A. Garcia, Saar Gill, Dylan M. Marchione, Konrad Gabrusiewicz, Kimberly Veliz, Xueqing Maggie Lu, Nicholas R. Anderson, Olga Shestova, Miriam Y. Kim, Feng Shen, Stephen R. Wallace, Michael Klichinsky, Xinhe Shan, Carl H. June, Roddy S. O’Connor, Kristin Blouch, Maksim Shestov, Marco Ruella, Yumi Yashiro-Ohtani, Miroslaw Kozlowski, Martha Zeeman, Katherine D. Cummins, Maggie Schmierer, Andrew Best, Nicholas E. Petty, and Saad S. Kenderian
- Subjects
Lung Neoplasms ,Cell Survival ,T cell ,Urology ,medicine.medical_treatment ,Antigen presentation ,Biomedical Engineering ,Bioengineering ,Biology ,Applied Microbiology and Biotechnology ,Immunotherapy, Adoptive ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,Cancer immunotherapy ,Cell Line, Tumor ,Neoplasms ,medicine ,Macrophage ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,Tumor microenvironment ,Microscopy, Video ,business.industry ,Macrophages ,Immunotherapy ,Neoplasms, Experimental ,Chimeric antigen receptor ,medicine.anatomical_structure ,Humanized mouse ,Cancer research ,Molecular Medicine ,business ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Chimeric antigen receptor (CAR) T cell therapy has shown promise in hematologic malignancies, but its application to solid tumors has been challenging1–4. Given the unique effector functions of macrophages and their capacity to penetrate tumors5, we genetically engineered human macrophages with CARs to direct their phagocytic activity against tumors. We found that a chimeric adenoviral vector overcame the inherent resistance of primary human macrophages to genetic manipulation and imparted a sustained pro-inflammatory (M1) phenotype. CAR macrophages (CAR-Ms) demonstrated antigen-specific phagocytosis and tumor clearance in vitro. In two solid tumor xenograft mouse models, a single infusion of human CAR-Ms decreased tumor burden and prolonged overall survival. Characterization of CAR-M activity showed that CAR-Ms expressed pro-inflammatory cytokines and chemokines, converted bystander M2 macrophages to M1, upregulated antigen presentation machinery, recruited and presented antigen to T cells and resisted the effects of immunosuppressive cytokines. In humanized mouse models, CAR-Ms were further shown to induce a pro-inflammatory tumor microenvironment and boost anti-tumor T cell activity. Primary macrophages engineered to express chimeric antigen receptors have anti-tumor activity in humanized mice.
- Published
- 2021
24. Histone H3.3 G34 mutations promote aberrant PRC2 activity and drive tumor progression
- Author
-
C. David Allis, Dylan M. Marchione, Shriya Deshmukh, Chao Lu, Nada Jabado, Benjamin A. Garcia, Siddhant U. Jain, Daniel N. Weinberg, Sima Khazaei, Nikoleta Juretic, Stefan M. Lundgren, Xiaoshi Wang, and Peter W. Lewis
- Subjects
Gene Expression ,Methylation ,Histones ,Histone H3 ,SETD2 ,Gene silencing ,Humans ,Neoplastic Processes ,Polycomb Repressive Complex 1 ,Multidisciplinary ,biology ,Lysine ,Polycomb Repressive Complex 2 ,Mesenchymal Stem Cells ,Glioma ,Histone-Lysine N-Methyltransferase ,Biological Sciences ,Chromatin ,Histone ,HEK293 Cells ,Gene Expression Regulation ,Tumor progression ,Mutation ,Cancer research ,biology.protein ,PRC2 ,Protein Processing, Post-Translational - Abstract
A high percentage of pediatric gliomas and bone tumors reportedly harbor missense mutations at glycine 34 in genes encoding histone variant H3.3. We find that these H3.3 G34 mutations directly alter the enhancer chromatin landscape of mesenchymal stem cells by impeding methylation at lysine 36 on histone H3 (H3K36) by SETD2, but not by the NSD1/2 enzymes. The reduction of H3K36 methylation by G34 mutations promotes an aberrant gain of PRC2-mediated H3K27me2/3 and loss of H3K27ac at active enhancers containing SETD2 activity. This altered histone modification profile promotes a unique gene expression profile that supports enhanced tumor development in vivo. Our findings are mirrored in G34W-containing giant cell tumors of bone where patient-derived stromal cells exhibit gene expression profiles associated with early osteoblastic differentiation. Overall, we demonstrate that H3.3 G34 oncohistones selectively promote PRC2 activity by interfering with SETD2-mediated H3K36 methylation. We propose that PRC2-mediated silencing of enhancers involved in cell differentiation represents a potential mechanism by which H3.3 G34 mutations drive these tumors.
- Published
- 2020
25. H3.3G34W promotes growth and impedes differentiation of osteoblast-like mesenchymal progenitors in Giant Cell Tumour of Bone
- Author
-
Jay S. Wunder, Jason Karamchandani, Ashot S. Harutyunyan, Nada Jabado, Robert E. Turcotte, Tianna S. Sihota, Damien Faury, Shriya Deshmukh, Peter W. Lewis, Kateryna Rossokhata, Stephen C. Mack, Brendan C. Dickson, Livia Garzia, Pierre Thibault, Leonie G. Mikael, Liam D. Hendrikse, Dylan M. Marchione, Carol C.L. Chen, Siddhant U. Jain, Takeaki Ishii, Sima Khazaei, Nicolas De Jay, Benjamin A. Garcia, Sungmi Jung, Véronique Lisi, Michael D. Taylor, Claudia L. Kleinman, Robert Eveleigh, Wajih Jawhar, Eric Bonneil, and Joel Lanoix
- Subjects
0301 basic medicine ,Stromal cell ,Population ,Gene Expression ,Bone Neoplasms ,Biology ,Article ,Extracellular matrix ,Histones ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Progenitor cell ,education ,Giant Cell Tumor of Bone ,education.field_of_study ,Tumor microenvironment ,Osteoblasts ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Osteoblast ,Cell Differentiation ,medicine.disease ,Chromatin ,Cell biology ,Nucleosomes ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Giant-cell tumor of bone - Abstract
Glycine 34-to-tryptophan (G34W) substitutions in H3.3 arise in approximately 90% of giant cell tumor of bone (GCT). Here, we show H3.3 G34W is necessary for tumor formation. By profiling the epigenome, transcriptome, and secreted proteome of patient samples and tumor-derived cells CRISPR–Cas9-edited for H3.3 G34W, we show that H3.3K36me3 loss on mutant H3.3 alters the deposition of the repressive H3K27me3 mark from intergenic to genic regions, beyond areas of H3.3 deposition. This promotes redistribution of other chromatin marks and aberrant transcription, altering cell fate in mesenchymal progenitors and hindering differentiation. Single-cell transcriptomics reveals that H3.3 G34W stromal cells recapitulate a neoplastic trajectory from a SPP1+ osteoblast-like progenitor population toward an ACTA2+ myofibroblast-like population, which secretes extracellular matrix ligands predicted to recruit and activate osteoclasts. Our findings suggest that H3.3 G34W leads to GCT by sustaining a transformed state in osteoblast-like progenitors, which promotes neoplastic growth, pathologic recruitment of giant osteoclasts, and bone destruction. Significance: This study shows that H3.3 G34W drives GCT tumorigenesis through aberrant epigenetic remodeling, altering differentiation trajectories in mesenchymal progenitors. H3.3 G34W promotes in neoplastic stromal cells an osteoblast-like progenitor state that enables undue interactions with the tumor microenvironment, driving GCT pathogenesis. These epigenetic changes may be amenable to therapeutic targeting in GCT. See related commentary by Licht, p. 1794. This article is highlighted in the In This Issue feature, p. 1775
- Published
- 2020
26. Chromatin-mediated alternative splicing regulates cocaine-reward behavior
- Author
-
Rachel L. Neve, Sonia I. Lombroso, Peter J. Hamilton, Song-Jun Xu, Marco D. Carpenter, Delaney K. Fischer, Mathieu E. Wimmer, Carissa J. Lim, R. Christopher Pierce, Dylan M. Marchione, Benjamin A. Garcia, and Elizabeth A. Heller
- Subjects
Male ,Spliceosome ,biology ,General Neuroscience ,Alternative splicing ,Self Administration ,Epigenome ,Chromatin ,Cell biology ,Epigenesis, Genetic ,Behavior, Addictive ,Mice, Inbred C57BL ,Alternative Splicing ,Mice ,Histone ,Cocaine ,Dopamine Uptake Inhibitors ,Reward ,RNA splicing ,biology.protein ,Animals ,Female ,Epigenetics ,Gene - Abstract
Neuronal alternative splicing is a key gene regulatory mechanism in the brain. However, the spliceosome machinery is insufficient to fully specify splicing complexity. In considering the role of the epigenome in activity-dependent alternative splicing, we and others find the histone modification H3K36me3 to be a putative splicing regulator. In this study, we found that mouse cocaine self-administration caused widespread differential alternative splicing, concomitant with the enrichment of H3K36me3 at differentially spliced junctions. Importantly, only targeted epigenetic editing can distinguish between a direct role of H3K36me3 in splicing and an indirect role via regulation of splice factor expression elsewhere on the genome. We targeted Srsf11, which was both alternatively spliced and H3K36me3 enriched in the brain following cocaine self-administration. Epigenetic editing of H3K36me3 at Srsf11 was sufficient to drive its alternative splicing and enhanced cocaine self-administration, establishing the direct causal relevance of H3K36me3 to alternative splicing of Srsf11 and to reward behavior.
- Published
- 2020
27. Chromatin-Mediated Alternative Splicing Regulates Cocaine Reward Behavior
- Author
-
Elizabeth A. Heller, Carissa J. Lim, Sonia I. Lombroso, Benjamin A. Garcia, Peter J. Hamilton, Marco D. Carpenter, R. Christopher Pierce, Mathieu E. Wimmer, Rachel L. Neve, Song-Jun Xu, Dylan M. Marchione, and Delaney K. Fischer
- Subjects
Regulation of gene expression ,Exon ,Histone ,biology ,RNA splicing ,Alternative splicing ,biology.protein ,Regulator ,Epigenetics ,Chromatin ,Cell biology - Abstract
Mechanisms by which epigenetic modifications regulate alternative splicing are largely unexplored. Differential alternative splicing and histone modification enrichment are key mechanisms for neuronal gene regulation. Both are grossly altered in mouse brain following investigator-administered cocaine. Our group and others have identified the histone modification, H3K36me3, as a putative splicing regulator. In the current study, we found that mouse cocaine self-administration caused widespread differential alternative splicing, concomitant with enrichment of H3K36me3 at differentially spliced junctions. The splice factor motif for Srsf11 was enriched in splice junctions of cocaine induced alternative exons, yet Srsf11 expression was unchanged by cocaine treatment. Rather, Srsf11 was both differentially spliced and enriched in H3K36me3. Epigenetic editing showed that H3K36me3 functions directly in alternative splicing of Srsf11. Finally, both Srsf11-targeted and global H3K36me3 enrichment enhanced cocaine self-administration. These findings established the direct causal relevance of H3K36me3 to alternative splicing of Srsf11 and to reward behavior.
- Published
- 2020
28. Take a Step Down and Beware of H3K36me2: The H3K27m Mutation in Glioma Directs H3K27 Methylation
- Author
-
Brian Krug, Tianyuan Lu, Benjamin A. Garcia, Eric Bareke, Caterina Russo, Nada Jabado, Haifen Chen, Mariel Coradin, Cynthia Horth, Dylan M. Marchione, Jacek Majewski, Hamid Nikbakht, and Ashot S. Harutyunyan
- Subjects
Mutation ,biology ,fungi ,Epigenome ,medicine.disease ,medicine.disease_cause ,Glioma ,Histone methylation ,medicine ,Cancer research ,biology.protein ,Gene silencing ,Cancer epigenetics ,PRC2 ,Epigenomics - Abstract
The discovery of H3K27M mutations in pediatric gliomas marked a new chapter in cancer epigenetics. Numerous studies have investigated the effect of this mutation on H3K27 trimethylation, but only recently have we started to realize its additional effects on the epigenome. Here, we use isogenic glioma H3K27M(+/-) cell lines to investigate H3K27 methylation and its interaction with H3K36 and H3K9 modifications. We describe a “Step Down” effect of H3K27M on the distribution of H3K27 methylation: me3 is reduced to me2, me2 is reduced to me1, while H3K36me2/3 delineate the boundaries for the spread of H3K27me marks. We also observe a replacement of H3K27me2/3 silencing by H3K9me3. Using a computational simulation, we explain our observations by reduced effectiveness of PRC2 and constraints imposed on deposition of H3K27me by antagonistic H3K36 modifications. Our work further elucidates the effect of H3K27M in gliomas, as well as general principles of deposition of H3K27 methylation.
- Published
- 2020
29. Histone H3.3 beyond cancer: Germline mutations in Histone 3 Family 3A and 3B cause a previously unidentified neurodegenerative disorder in 46 patients
- Author
-
Thomas Besnard, Kristian Tveten, Hilary F Kitson, Jennifer A. Lee, Brieana Fregeau, Rachel Schot, Khadija Wilson, Katrin Õunap, Juliane Winkelmann, Anna Lehman, Nicola Longo, Servi J. C. Stevens, Megan T. Cho, Christina G.S. Palmer, Causes Study, Giovanni Battista Ferrero, Joy Dean, Lone W. Laulund, Grazia M.S. Mancini, Matias Wagner, Martin G. Martin, Sabine Lüttgen, Elizabeth J. Bhoj, Amanda J. Yoon, Thomas Klopstock, Janet S. Sinsheimer, Eric Vilain, Sébastien Küry, Francesca Clementina Radio, Jiddeke M. van de Kamp, Cameron Mrokse, Hakon Hakonarson, Samuel G. Cox, Jeanette C. Papp, Margot I. Van Allen, Raymond J. Louie, Constance T. R. M. Stumpel, Evan F. Joiner, Juanita Neira, Arve Vøllo, Amy Pizzino, Kelly Radtke, Celeste Simon, Michelle L. Thompson, Allison Zheng, Omar Sherbini, Marcia C. Willing, Tim M. Strom, Benjamin Garcia, Sara S. Cathey, Theresa A. Grebe, Dong Li, Marjan M. Weiss, Marco Tartaglia, Laura M Bryant, Sandra Mercier, Katherine L. Helbig, Martin Jakob Larsen, Ddd Study, Alexandrea Wadley, Alexander P.A. Stegmann, Sabina Barresi, A. Micheil Innes, Elaine H. Zackai, Gregory Costain, Davor Lessel, Molly Snyder, Heather P. Crawford, Richard Redon, Pearl Lee, Melissa Byler, Holly Dubbs, J. Gage Crump, K. E. Stuurman, Boris Keren, Stéphane Bézieau, Stan F. Nelson, Kristin G. Monaghan, Michael J. Lyons, Jeffrey W. Innis, Anna C.E. Hurst, Elizabeth A. Sellars, Samantha A. Schrier Vergano, Saadet Mercimek-Andrews, Monica H. Wojcik, Alison Ross, Heiko Reutter, Zuo-Fei Yuan, Dylan M. Marchione, Renee Bend, Diana Carli, Zöe Powis, Neil H. Parker, Jennifer Muncy Thomas, Luis A. Umaña, Adeline Vanderver, Julia Hoefele, Linda Manwaring, Christina Fagerberg, Elly Brokamp, M. Stephen Meyn, Pilvi Ilves, Xavier de la Cruz, Nina Powell-Hamilton, Caroline Nava, Garrett Gotway, Karit Reinson, Kristin D. Kernohan, Jennifer Norman, Alexandra Afenjar, Benjamin Cogné, Delphine Héron, Roman Günthner, Alfredo Brusco, John Dean, Kevin A. Janssen, Robert Roger Lebel, Divya Nair, Jijun Wan, Julian A. Martinez-Agosto, Elliott H. Sherr, Kyle Retterer, Claudia B. Catarino, Michael E. March, Natalia Padilla, Elise Brimble, Sylvie Odent, Jane L. Schuette, David Chitayat, Klaas J. Wierenga, Kirsty McWalter, Trine Prescott, Jonas Denecke, Wendy K. Chung, Human genetics, Amsterdam Neuroscience - Complex Trait Genetics, Amsterdam Gastroenterology Endocrinology Metabolism, Klinische Genetica, MUMC+: DA KG Polikliniek (9), RS: GROW - R4 - Reproductive and Perinatal Medicine, MUMC+: DA KG Lab Centraal Lab (9), and Clinical Genetics
- Subjects
metabolism [Zebrafish Proteins] ,RESIDUE ,metabolism [Histones] ,GENES ,Somatic cell ,CODE ,cancer mutation ,histone ,Biology ,VARIANTS ,medicine.disease_cause ,progressive neurologic dysfunction ,Histones ,03 medical and health sciences ,Histone H3 ,0302 clinical medicine ,Germline mutation ,SDG 3 - Good Health and Well-being ,histone, neurodevelopmental disorder, progressive neurologic dysfunction, congenital anomalies, cancer mutation ,medicine ,Animals ,Humans ,H3-3A protein, human ,metabolism [Zebrafish] ,TRANSCRIPTION ,PHOSPHORYLATION ,Gene ,Zebrafish ,Germ-Line Mutation ,030304 developmental biology ,Genetics ,genetics [Zebrafish] ,0303 health sciences ,Multidisciplinary ,foxd3 protein, zebrafish ,congenital anomalies ,Forkhead Transcription Factors ,Zebrafish Proteins ,biology.organism_classification ,genetics [Histones] ,neurodevelopmental disorder ,H3F3B ,Histone ,genetics [Forkhead Transcription Factors] ,genetics [Neurodegenerative Diseases] ,biology.protein ,ddc:500 ,Carcinogenesis ,030217 neurology & neurosurgery - Abstract
Germ line mutations in H3F3A and H3F3B cause a previously unidentified neurodevelopmental syndrome. Although somatic mutations in Histone 3.3 (H3.3) are well-studied drivers of oncogenesis, the role of germline mutations remains unreported. We analyze 46 patients bearing de novo germline mutations in histone 3 family 3A (H3F3A) or H3F3B with progressive neurologic dysfunction and congenital anomalies without malignancies. Molecular modeling of all 37 variants demonstrated clear disruptions in interactions with DNA, other histones, and histone chaperone proteins. Patient histone posttranslational modifications (PTMs) analysis revealed notably aberrant local PTM patterns distinct from the somatic lysine mutations that cause global PTM dysregulation. RNA sequencing on patient cells demonstrated up-regulated gene expression related to mitosis and cell division, and cellular assays confirmed an increased proliferative capacity. A zebrafish model showed craniofacial anomalies and a defect in Foxd3-derived glia. These data suggest that the mechanism of germline mutations are distinct from cancer-associated somatic histone mutations but may converge on control of cell proliferation
- Published
- 2020
30. Proteomic approaches for cancer epigenetics research
- Author
-
John Wojcik, Dylan M. Marchione, and Benjamin A. Garcia
- Subjects
Proteomics ,0301 basic medicine ,Computational biology ,medicine.disease_cause ,Biochemistry ,Article ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,Neoplasms ,medicine ,Humans ,Histone code ,Cancer epigenetics ,Epigenetics ,Molecular Biology ,030102 biochemistry & molecular biology ,biology ,Chromatin ,030104 developmental biology ,Histone ,Cancer cell ,biology.protein ,Carcinogenesis - Abstract
Introduction: Epigenetic dysregulation drives or supports numerous human cancers. The chromatin landscape in cancer cells is often marked by abnormal histone post-translational modification (PTM) patterns and by aberrant assembly and recruitment of protein complexes to specific genomic loci. Mass spectrometry-based proteomic analyses can support the discovery and characterization of both phenomena. Areas covered: We broadly divide this literature into two parts: 'modification-centric' analyses that link histone PTMs to cancer biology; and 'complex-centric' analyses that examine protein-protein interactions that occur de novo as a result of oncogenic mutations. We also discuss proteomic studies of oncohistones. We highlight relevant examples, discuss limitations, and speculate about forthcoming innovations regarding each application. Expert commentary: 'Modification-centric' analyses have been used to further understanding of cancer's histone code and to identify associated therapeutic vulnerabilities. 'Complex-centric' analyses have likewise revealed insights into mechanisms of oncogenesis and suggested potential therapeutic targets, particularly in MLL-associated leukemia. Proteomic experiments have also supported some of the pioneering studies of oncohistone-mediated tumorigenesis. Additional applications of proteomics that may benefit cancer epigenetics research include middle-down and top-down histone PTM analysis, chromatin reader profiling, and genomic locus-specific protein identification. In the coming years, proteomic approaches will remain powerful ways to interrogate the biology of cancer.
- Published
- 2018
31. Chromatin-mediated alternative splicing regulates cocaine reward behavior
- Author
-
Dylan M. Marchione, Elizabeth A. Heller, Carissa J. Lim, Peter J. Hamilton, Marco D. Carpenter, Rachel L. Neve, Song-Jun Xu, and Sonia I. Lombroso
- Subjects
Regulation of gene expression ,Histone ,biology ,Transcription (biology) ,RNA splicing ,Alternative splicing ,biology.protein ,Regulator ,Epigenetics ,Chromatin ,Cell biology - Abstract
Alternative splicing is a key mechanism for neuronal gene regulation, and is grossly altered in mouse brain reward regions following investigator-administered cocaine. It is well established that cocaine epigenetically regulates transcription, yet mechanism(s) by which cocaine-induced epigenetic modifications regulate alternative splicing is largely unexplored. Our group and others have previously identified the histone modification, H3K36me3, as a putative splicing regulator. However, it has not yet been possible to establish the direct causal relevance of this modification to alternative splicing in brain or any other context. We found that mouse cocaine self-administration caused widespread alternative splicing, concomitant with enrichment of H3K36me3 at splice junctions. Differentially spliced genes were enriched in the motif for splice factor, Srsf11, which was both differentially spliced and enriched in H3K36me3. Epigenetic editing led us to conclude that H3K36me3 functions directly in alternative splicing of Srsf11, and that Set2 mediated H3K36me3 bidirectionally regulates cocaine intake.
- Published
- 2019
- Full Text
- View/download PDF
32. HYPERsol: flash-frozen results from archival FFPE tissue for clinical proteomics
- Author
-
John Wojcik, John P. Wilson, Ilyana Ilieva, Benjamin A. Garcia, Darryl J. Pappin, and Dylan M. Marchione
- Subjects
0303 health sciences ,03 medical and health sciences ,0302 clinical medicine ,Formalin fixed paraffin embedded ,Solubilization ,Computer science ,030220 oncology & carcinogenesis ,Protein purification ,Proteome ,Sample processing ,Computational biology ,Proteomics ,030304 developmental biology - Abstract
Massive formalin-fixed, paraffin-embedded (FFPE) tissue archives exist worldwide, representing a potential gold mine for clinical proteomics research. However, current protocols for FFPE proteomics lack standardization, efficiency, reproducibility, and scalability. Here we present High-Yield Protein Extraction and Recovery by direct SOLubilization (HYPERsol), an optimized workflow using adaptive-focused acoustics (AFA) ultrasonication and S-Trap sample processing that enables proteome coverage and quantification from FFPE samples comparable to that achieved from flash-frozen tissue (average R = 0.936).
- Published
- 2019
33. Histone H3.3G34-Mutant Interneuron Progenitors Co-opt PDGFRA for Gliomagenesis
- Author
-
Mathieu Blanchette, Albert M. Berghuis, Hiromichi Suzuki, Pratiti Bandopadhayay, Dong Anh Khuong-Quang, Dylan M. Marchione, Nicolas De Jay, Wajih Jawhar, Angelia V. Bassenden, Djihad Hadjadj, Ashot S. Harutyunyan, Shriya Deshmukh, Steffen Albrecht, Michele Zeinieh, Nikoleta Juretic, Paolo Salomoni, Katerina Vanova, Ales Vicha, Stefan M. Pfister, Manav Pathania, Selin Jessa, Almos Klekner, Leonie G. Mikael, CM Kramm, David T.W. Jones, Tenzin Gayden, Sebastian Brandner, Michal Zapotocky, Nicola Maestro, Eleanor Woodward, Alexander G. Weil, David S. Ziegler, Jordan R. Hansford, Steven Hébert, Frank Dubois, Benjamin Ellezam, Deli A, Damien Faury, Véronique Lisi, Augusto Faria Andrade, Andrey Korshunov, Mariella G. Filbin, Michael D. Taylor, Claudia L. Kleinman, Andrea Bajic, Carol C.L. Chen, Caterina Russo, Nada Jabado, Peter Hauser, Benjamin A. Garcia, Stephen C. Mack, Keith L. Ligon, David Sumerauer, Lenka Krskova, Jason Karamchandani, Rameen Beroukhim, Rola Dali, László Bognár, Dominik Sturm, József Virga, Marie Coutelier, Livia Garzia, Paul G Ekert, and Josef Zamecnik
- Subjects
genetics [Glioma] ,metabolism [Histones] ,Receptor, Platelet-Derived Growth Factor alpha ,Transcription, Genetic ,Carcinogenesis ,pathology [Carcinogenesis] ,genetics [Transcriptome] ,metabolism [Neural Stem Cells] ,medicine.disease_cause ,Epigenesis, Genetic ,Histones ,chromatin conformation ,0302 clinical medicine ,Neural Stem Cells ,genetics [Carcinogenesis] ,Promoter Regions, Genetic ,metabolism [Interneurons] ,pathology [Astrocytes] ,0303 health sciences ,Mutation ,metabolism [Astrocytes] ,biology ,Brain Neoplasms ,cell-of-origin ,Glioma ,metabolism [Receptor, Platelet-Derived Growth Factor alpha] ,Cellular Reprogramming ,genetics [Histones] ,metabolism [Lysine] ,Chromatin ,pediatric cancer ,Gene Expression Regulation, Neoplastic ,Oligodendroglia ,genetics [Cellular Reprogramming] ,PDGFRA ,Histone ,GSX2 ,Lineage (genetic) ,pathology [Brain Neoplasms] ,interneuron progenitors ,metabolism [Chromatin] ,genetics [Mutation] ,Context (language use) ,embryology [Prosencephalon] ,Models, Biological ,Article ,General Biochemistry, Genetics and Molecular Biology ,metabolism [Oligodendroglia] ,H3.3 G34R/V ,03 medical and health sciences ,Histone H3 ,Prosencephalon ,Interneurons ,medicine ,Animals ,Cell Lineage ,ddc:610 ,Gene Silencing ,metabolism [Embryo, Mammalian] ,030304 developmental biology ,Lysine ,single-cell transcriptome ,Embryo, Mammalian ,Pediatric cancer ,oncohistones ,digestive system diseases ,genetics [Receptor, Platelet-Derived Growth Factor alpha] ,genetics [Brain Neoplasms] ,Mice, Inbred C57BL ,gliomas ,Astrocytes ,genetics [Promoter Regions, Genetic] ,biology.protein ,Cancer research ,Neoplasm Grading ,Transcriptome ,pathology [Glioma] ,030217 neurology & neurosurgery - Abstract
Histone H3.3 glycine 34 to arginine/valine (G34R/V) mutations drive deadly gliomas and show exquisite regional and temporal specificity, suggesting a developmental context permissive to their effects. Here, we show that 50% of G34R/V-tumours (n=95) bear activating PDGFRA mutations that display strong selection pressure at recurrence. While considered gliomas, G34R/V-tumours actually arise in GSX2/DLX-expressing interneuron progenitors, where G34R/V-mutations impair neuronal differentiation. The lineage-of-origin may facilitate PDGFRA co-option through a chromatin loop connecting PDGFRA to GSX2 regulatory elements, promoting PDGFRA overexpression and mutation. At the single-cell level, G34R/V-tumours harbour dual neuronal/astroglial identity and lack oligodendroglial programs, actively repressed by GSX2/DLX-mediated cell-fate specification. G34R/V may become dispensable for tumour maintenance, while mutant-PDGFRA is potently oncogenic. Collectively, our results open novel research avenues in deadly tumours. G34R/V-gliomas are neuronal malignancies, where interneuron progenitors are stalled in differentiation by G34R/V-mutations, and malignant gliogenesis is promoted by co-option of a potentially targetable pathway, PDGFRA signalling.
- Published
- 2020
34. Glutathione antioxidant pathway activity and reserve determine toxicity and specificity of the biliary toxin biliatresone in zebrafish
- Author
-
Donghun Shin, John R. Porter, Kyung A. Koo, Rebecca G. Wells, Juhoon So, Kevin P. Gillespie, Kristin Lorent, Xiao Zhao, Dylan M. Marchione, Ian A. Blair, Orith Waisbourd-Zinman, Michael Pack, and Benjamin J. Wilkins
- Subjects
0301 basic medicine ,Antioxidant ,Hepatology ,biology ,medicine.medical_treatment ,Glutathione ,biology.organism_classification ,Cholangiocyte ,Biliatresone ,Cell biology ,Acetylcysteine ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,medicine.anatomical_structure ,Biochemistry ,chemistry ,Hepatocyte ,Toxicity ,medicine ,Zebrafish ,medicine.drug - Abstract
Biliatresone is an electrophilic isoflavone isolated from Dysphania species plants that has been causatively linked to naturally occurring outbreaks of a biliary atresia (BA)-like disease in livestock. Biliatresone has selective toxicity for extrahepatic cholangiocytes (EHCs) in zebrafish larvae. To better understand its mechanism of toxicity, we performed transcriptional profiling of liver cells isolated from zebrafish larvae at the earliest stage of biliatresone-mediated biliary injury, with subsequent comparison of biliary and hepatocyte gene expression profiles. Transcripts encoded by genes involved in redox stress response, particularly those involved in glutathione (GSH) metabolism, were among the most prominently up-regulated in both cholangiocytes and hepatocytes of biliatresone-treated larvae. Consistent with these findings, hepatic GSH was depleted at the onset of biliary injury, and in situ mapping of the hepatic GSH redox potential using a redox-sensitive green fluorescent protein biosensor showed that it was significantly more oxidized in EHCs both before and after treatment with biliatresone. Pharmacological and genetic manipulation of GSH redox homeostasis confirmed the importance of GSH in modulating biliatresone-induced injury given that GSH depletion sensitized both EHCs and the otherwise resistant intrahepatic cholangiocytes to the toxin, whereas replenishing GSH level by N-acetylcysteine administration or activation of nuclear factor erythroid 2-like 2 (Nrf2), a transcriptional regulator of GSH synthesis, inhibited EHC injury. Conclusion: These findings strongly support redox stress as a critical contributing factor in biliatresone-induced cholangiocyte injury, and suggest that variations in intrinsic stress responses underlie the susceptibility profile. Insufficient antioxidant capacity of EHCs may be critical to early pathogenesis of human BA. (Hepatology 2016;64:894-907)
- Published
- 2016
35. H3K27M in Gliomas Causes a One-Step Decrease in H3K27 Methylation and Reduced Spreading within the Constraints of H3K36 Methylation
- Author
-
Hamid Nikbakht, Tianyuan Lu, Dylan M. Marchione, Brian Krug, Mariel Coradin, Benjamin A. Garcia, Nada Jabado, Ashot S. Harutyunyan, Caterina Russo, Eric Bareke, Jacek Majewski, Cynthia Horth, and Haifen Chen
- Subjects
computational modeling ,Epigenomics ,0301 basic medicine ,Gene Expression ,Biology ,medicine.disease_cause ,Methylation ,Article ,General Biochemistry, Genetics and Molecular Biology ,Histones ,03 medical and health sciences ,Methionine ,0302 clinical medicine ,Cell Line, Tumor ,Glioma ,Histone methylation ,medicine ,Humans ,Gene silencing ,histone methylation ,lcsh:QH301-705.5 ,Mutation ,H3.3K27M ,Lysine ,fungi ,Polycomb Repressive Complex 2 ,Epigenome ,DNA Methylation ,medicine.disease ,Chromatin ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,lcsh:Biology (General) ,pediatric high-grade glioma ,Cancer research ,biology.protein ,PRC2 ,Protein Processing, Post-Translational ,030217 neurology & neurosurgery - Abstract
SUMMARY The discovery of H3K27M mutations in pediatric gliomas marked a new chapter in cancer epigenomics. Numerous studies have investigated the effect of this mutation on H3K27 trimethylation, but only recently have we started to realize its additional effects on the epigenome. Here, we use isogenic glioma H3K27M+/− cell lines to investigate H3K27 methylation and its interaction with H3K36 and H3K9 modifications. We describe a “step down” effect of H3K27M on the distribution of H3K27 methylation: me3 is reduced to me2, me2 is reduced to me1, whereas H3K36me2/3 delineates the boundaries for the spread of H3K27me marks. We also observe a replacement of H3K27me2/3 silencing by H3K9me3. Using a computational simulation, we explain our observations by reduced effectiveness of PRC2 and constraints imposed on the deposition of H3K27me by antagonistic H3K36 modifications. Our work further elucidates the effects of H3K27M in gliomas as well as the general principles of deposition in H3K27 methylation., Graphical Abstract, In Brief Harutyunyan et al. use isogenic glioma H3K27M+/− cell lines to demonstrate the rewiring of the epigenome, specifically H3K27me1/2/3, H3K36me2/3, and H3K9me3. The dynamic deposition of histone marks is simulated by a stochastic model. This work further advances the understanding of the deposition of H3K27 methylation in H3K27M mutant gliomas.
- Published
- 2020
36. Abstract PR07: Human chimeric antigen receptor (CAR) macrophages for cancer immunotherapy
- Author
-
Miroslaw Kozlowski, Kristin Blouch, Roddy S. O’Connor, Benjamin A. Garcia, Andrew Best, Michael Klichinsky, Stephen R. Wallace, Saar Gill, Dylan M. Marchione, Maksim Shestov, Xueqing Maggie Lu, Miriam Y. Kim, Olga Shestova, Carl H. June, Marco Ruella, and Saad S. Kenderian
- Subjects
Cancer Research ,Tumor microenvironment ,business.industry ,medicine.medical_treatment ,Immunology ,Immunotherapy ,Chimeric antigen receptor ,Cell therapy ,Immune system ,Cancer immunotherapy ,Antigen ,Interferon ,Cancer research ,Medicine ,business ,medicine.drug - Abstract
Despite recent landmark advances in T-cell immunotherapy for the treatment of human cancer, metastatic solid tumors remain an intractable challenge. Macrophages are often the most abundant immune cell in the tumor microenvironment (TME), where they may convert into immunosuppressive (M2) tumor-associated macrophages (TAMs) and participate in disease progression. Currently, macrophage-orientated immunotherapeutic approaches under clinical development in oncology seek to reduce TAM infiltration (CSF-1 antagonists) or enhance TAM phagocytosis (CD47 antagonists). Transfer of autologous, activated, but nontargeted macrophages failed to demonstrate antitumor efficacy in past clinical trials. We hypothesized that genetically engineering human macrophages with CARs against tumor-associated antigens could redirect their phagocytic activity and lead to therapeutic efficacy with the potential for the induction of an antitumor T-cell response. We first demonstrate that CD3-zeta-based CARs are capable of inducing phagocytosis in human THP-1 macrophages, while truncated intracellular-domain deficient CARs were not. Targeted phagocytosis and clearance of CD19+, mesothelin +, and HER2+ cells by CARs targeted against each respective antigen was significantly superior to that by control untransduced (UTD) macrophages. We demonstrate that primary human macrophages, which are resistant to most viral vectors, are efficiently transduced by the chimeric fiber adenoviral vector Ad5f35 (~70% in 10 normal donors). Using Ad5f35, we engineered primary human macrophages with a CD3-zeta-based CAR against HER2. Anti-HER2 primary human CAR macrophages demonstrated targeted phagocytosis against HER2+ but not HER2- cell lines, with phagocytic activity dependent on both the CAR and antigen densities. Furthermore, CAR, but not UTD, macrophages led to potent dose-dependent killing of three distinct HER2-high cell lines in vitro. We sought to test the efficacy of anti-HER2 primary human macrophages in xenograft models of human HER2+ ovarian cancer. A single dose of CAR, but not UTD macrophages, led to tumor regression and improved overall survival in both intraperitoneal and disseminated models of disease. We show that macrophage transduction with Ad5f35, a double-stranded DNA virus, leads to a broad gene expression change, an interferon signaling signature, and phenotypic clustering toward classically activated M1 macrophages. CAR macrophages upregulated co-stimulatory ligand and antigen processing/presentation genes and led to enhanced T-cell stimulation in vitro and in vivo. Lastly, CAR, but not UTD, macrophages showed a broad resistance for M2 conversion in response to immunosuppressive cytokines. In conclusion, we show that primary human CAR macrophages are capable of targeted tumor phagocytosis, lead to improved overall survival in xenograft models, and demonstrate enhanced T-cell stimulation. CAR macrophages are a novel cell therapy platform for the treatment of human cancer. This abstract is also being presented as Poster B29. Citation Format: Michael Klichinsky, Marco Ruella, Olga Shestova, Andrew Best, Kristin Blouch, Xueqing M. Lu, Saad S. Kenderian, Miriam Y. Kim, Roddy O'Connor, Stephen Wallace, Miroslaw Kozlowski, Dylan M. Marchione, Maksim Shestov, Benjamin A. Garcia, Carl June, Saar Gill. Human chimeric antigen receptor (CAR) macrophages for cancer immunotherapy [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27-30; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(4 Suppl):Abstract nr PR07.
- Published
- 2020
37. Epigenomic Reordering Induced by Polycomb Loss Drives Oncogenesis but Leads to Therapeutic Vulnerabilities in Malignant Peripheral Nerve Sheath Tumors
- Author
-
Simone Sidoli, John Wojcik, Jacek Majewski, Amanda Lisby, Anissa Djedid, Dylan M. Marchione, and Benjamin A. Garcia
- Subjects
0301 basic medicine ,Epigenomics ,Proteomics ,Cancer Research ,Carcinogenesis ,Malignant peripheral nerve sheath tumor ,macromolecular substances ,medicine.disease_cause ,Nerve Sheath Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,biology ,medicine.disease ,030104 developmental biology ,Histone ,Oncology ,Neurofibrosarcoma ,030220 oncology & carcinogenesis ,DNA methylation ,biology.protein ,Cancer research ,Histone deacetylase ,PRC2 ,Nerve sheath neoplasm - Abstract
Malignant peripheral nerve sheath tumor (MPNST) is an aggressive sarcoma with recurrent loss-of-function alterations in polycomb-repressive complex 2 (PRC2), a histone-modifying complex involved in transcriptional silencing. To understand the role of PRC2 loss in pathogenesis and identify therapeutic targets, we conducted parallel global epigenomic and proteomic analysis of archival formalin-fixed, paraffin-embedded (FFPE) human MPNST with and without PRC2 loss (MPNSTLOSS vs. MPNSTRET). Loss of PRC2 resulted in increased histone posttranslational modifications (PTM) associated with active transcription, most notably H3K27Ac and H3K36me2, whereas repressive H3K27 di- and trimethylation (H3K27me2/3) marks were globally lost without a compensatory gain in other repressive PTMs. Instead, DNA methylation globally increased in MPNSTLOSS. Epigenomic changes were associated with upregulation of proteins in growth pathways and reduction in IFN signaling and antigen presentation, suggesting a role for epigenomic changes in tumor progression and immune evasion, respectively. These changes also resulted in therapeutic vulnerabilities. Knockdown of NSD2, the methyltransferase responsible for H3K36me2, restored MHC expression and induced interferon pathway expression in a manner similar to PRC2 restoration. MPNSTLOSS were also highly sensitive to DNA methyltransferase and histone deacetylase (HDAC) inhibitors. Overall, these data suggest that global loss of PRC2-mediated repression renders MPNST differentially dependent on DNA methylation to maintain transcriptional integrity and makes them susceptible to therapeutics that promote aberrant transcription initiation. Significance: Global profiling of histone PTMs and protein expression in archival human MPNST illustrates how PRC2 loss promotes oncogenesis but renders tumors vulnerable to pharmacologic modulation of transcription. See related commentary by Natarajan and Venneti, p. 3172
- Published
- 2018
38. H3K27M induces defective chromatin spread of PRC2-mediated repressive H3K27me2/me3 and is essential for glioma tumorigenesis
- Author
-
Gael Cagnone, Siddhant U. Jain, Warren A. Cheung, Jacek Majewski, Simon Papillon-Cavanagh, Shriya Deshmukh, Hamid Nikbakht, Jad I. Belle, Haifen Chen, Damien Faury, Benjamin Ellezam, Peter W. Lewis, Carol C.L. Chen, Nicolas De Jay, Abdulshakour Mohammadnia, Bo Hu, Melissa K. McConechy, Brian Krug, Dylan M. Marchione, Claudia L. Kleinman, Michele Zeinieh, Chao Lu, Ashot S. Harutyunyan, Tomi Pastinen, Leonie G. Mikael, Benjamin A. Garcia, Manav Pathania, Alexander G. Weil, Nada Jabado, Rui Li, Alexandre Montpetit, Denise Bechet, and Paolo Salomoni
- Subjects
0301 basic medicine ,Male ,metabolism [Histones] ,Carcinogenesis ,metabolism [Polycomb Repressive Complex 2] ,genetics [Histone Code] ,General Physics and Astronomy ,02 engineering and technology ,Mice, SCID ,medicine.disease_cause ,genetics [Glioblastoma] ,Epigenesis, Genetic ,pathology [Glioblastoma] ,Histones ,Mice ,Methionine ,Mice, Inbred NOD ,genetics [Carcinogenesis] ,Histone code ,lcsh:Science ,Child ,Regulation of gene expression ,Gene Editing ,Multidisciplinary ,biology ,Brain Neoplasms ,Neurogenesis ,Polycomb Repressive Complex 2 ,021001 nanoscience & nanotechnology ,genetics [Histones] ,Chromatin ,3. Good health ,Cell biology ,genetics [Methionine] ,Gene Expression Regulation, Neoplastic ,Histone Code ,Histone ,genetics [Neurogenesis] ,DNA methylation ,Female ,ddc:500 ,0210 nano-technology ,PRC2 ,methods [Gene Editing] ,pathology [Brain Neoplasms] ,Adolescent ,metabolism [Chromatin] ,Science ,macromolecular substances ,genetics [DNA Methylation] ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,genetics [Lysine] ,Aged ,Cell Proliferation ,Lysine ,General Chemistry ,DNA Methylation ,Xenograft Model Antitumor Assays ,genetics [Brain Neoplasms] ,030104 developmental biology ,HEK293 Cells ,Mutation ,biology.protein ,lcsh:Q ,CpG Islands ,CRISPR-Cas Systems ,Glioblastoma ,genetics [Cell Proliferation] ,genetics [CpG Islands] - Abstract
Lys-27-Met mutations in histone 3 genes (H3K27M) characterize a subgroup of deadly gliomas and decrease genome-wide H3K27 trimethylation. Here we use primary H3K27M tumor lines and isogenic CRISPR-edited controls to assess H3K27M effects in vitro and in vivo. We find that whereas H3K27me3 and H3K27me2 are normally deposited by PRC2 across broad regions, their deposition is severely reduced in H3.3K27M cells. H3K27me3 is unable to spread from large unmethylated CpG islands, while H3K27me2 can be deposited outside these PRC2 high-affinity sites but to levels corresponding to H3K27me3 deposition in wild-type cells. Our findings indicate that PRC2 recruitment and propagation on chromatin are seemingly unaffected by K27M, which mostly impairs spread of the repressive marks it catalyzes, especially H3K27me3. Genome-wide loss of H3K27me3 and me2 deposition has limited transcriptomic consequences, preferentially affecting lowly-expressed genes regulating neurogenesis. Removal of H3K27M restores H3K27me2/me3 spread, impairs cell proliferation, and completely abolishes their capacity to form tumors in mice., Lysine27-to-methionine mutations in histone H3 genes (H3K27M) occur in a subgroup of gliomas and decrease genome-wide H3K27 trimethylation. Here the authors utilise primary H3K27M tumour lines and isogenic CRISPR-edited controls and show that H3K27M induces defective chromatin spread of PRC2-mediated repressive H3K27me2/me3.
- Published
- 2018
39. Pervasive H3K27 Acetylation Leads to ERV Expression and a Therapeutic Vulnerability in H3K27M Gliomas
- Author
-
Benjamin Ellezam, Paul Guilhamon, Peter W. Lewis, Nicolas De Jay, Nada Jabado, Josie Ursini-Siegel, Sameer Agnihotri, Mathieu Lupien, Peter B. Dirks, Paul Lasko, Ashot S. Harutyunyan, Stephen C. Mack, Damien Faury, Robert F. Koncar, Carol C.L. Chen, Paolo Salomoni, Dylan M. Marchione, Shriya Deshmukh, Daniel D. De Carvalho, Leonie G. Mikael, Alexander G. Weil, Claudia L. Kleinman, Melissa K. McConechy, Brian Krug, Kelsey C. Bertrand, Benjamin A. Garcia, Sima Khazaei, and Cheryl H. Arrowsmith
- Subjects
0301 basic medicine ,Epigenomics ,genetics [Glioma] ,Cancer Research ,metabolism [Histones] ,drug effects [Gene Expression Regulation, Neoplastic] ,Vulnerability ,medicine.disease_cause ,metabolism [Glioma] ,Histones ,0302 clinical medicine ,drug therapy [Brain Neoplasms] ,methods [Epigenomics] ,therapeutic use [Histone Deacetylase Inhibitors] ,Mutation ,0303 health sciences ,Brain Neoplasms ,Acetylation ,Glioma ,genetics [Histones] ,Chromatin ,Cell biology ,metabolism [Brain Neoplasms] ,3. Good health ,Gene Expression Regulation, Neoplastic ,Histone ,Enhancer Elements, Genetic ,Oncology ,Expression (architecture) ,030220 oncology & carcinogenesis ,metabolism [Chromatin] ,Biology ,Article ,03 medical and health sciences ,Cell Line, Tumor ,drug therapy [Glioma] ,medicine ,Humans ,ddc:610 ,Enhancer ,030304 developmental biology ,Cell Biology ,drug effects [Enhancer Elements, Genetic] ,genetics [Brain Neoplasms] ,Histone Deacetylase Inhibitors ,030104 developmental biology ,DNA demethylation ,Cancer cell ,biology.protein ,Cancer research ,Histone deacetylase ,pharmacology [Histone Deacetylase Inhibitors] - Abstract
High-grade gliomas (HGG) defined by histone 3 K27M driver mutations exhibit global loss of H3K27 trimethylation and reciprocal gain of H3K27 acetylation, respectively shaping repressive and active chromatin landscapes. We generated tumor-derived isogenic models bearing this mutation and show that it leads to pervasive H3K27ac deposition across the genome. In turn, active enhancers and promoters are not created de novo and instead reflect the epigenomic landscape of the cell of origin. H3K27ac is enriched at repeat elements, resulting in their increased expression, which in turn can be further amplified by DNA demethylation and histone deacetylase inhibitors providing an exquisite therapeutic vulnerability. These agents may therefore modulate anti-tumor immune responses as a therapeutic modality for this untreatable disease.
- Published
- 2018
40. EpiProfile 2.0: A Computational Platform for Processing Epi-Proteomics Mass Spectrometry Data
- Author
-
Zuo-Fei Yuan, Johayra Simithy, Dylan M. Marchione, Benjamin A. Garcia, Simone Sidoli, Mary R. Szurgot, and Kevin A. Janssen
- Subjects
0301 basic medicine ,Epigenomics ,Proteomics ,Computational biology ,Mass spectrometry ,Biochemistry ,Article ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Tandem Mass Spectrometry ,Humans ,Epigenetics ,biology ,General Chemistry ,Chromatin ,Peak detection ,030104 developmental biology ,Histone ,030220 oncology & carcinogenesis ,DNA methylation ,biology.protein ,Retention time ,Protein Processing, Post-Translational ,Software ,Chromatography, Liquid ,HeLa Cells - Abstract
Epigenetics has become a fundamental scientific discipline with various implications for biology and medicine. Epigenetic marks, mostly DNA methylation and histone post-translational modifications (PTMs), play important roles in chromatin structure and function. Accurate quantification of these marks is an ongoing challenge due to the variety of modifications and their wide dynamic range of abundance. Here, we present EpiProfile 2.0, an extended version of our 2015 software (v1.0) for accurate quantification of histone peptides based on liquid chromatography – tandem mass spectrometry (LC-MS/MS) analysis. EpiProfile 2.0 is now optimized for data-independent acquisition through the use of precursor and fragment extracted ion chromatography to accurately determine the chromatographic profile and to discriminate isobaric forms of peptides. The software uses an intelligent retention time prediction trained on the analyzed samples to enable accurate peak detection. EpiProfile 2.0 supports label-free and isotopic labeling, different organisms, known sequence mutations in diseases, different derivatization strategies, and unusual PTMs (such as acyl-derived modifications). In summary, EpiProfile 2.0 is a universal and accurate platform for the quantification of histone marks via LC-MS/MS. Being the first software of its kind we anticipate that EpiProfile 2.0 will play a fundamental role in epigenetic studies relevant to biology and translational medicine. EpiProfile is freely available at https://github.com/zfyuan/EpiProfile2.0_Family.
- Published
- 2018
41. LC-MS Analysis of Human Platelets as a Platform for Studying Mitochondrial Metabolism
- Author
-
Qingqing Wang, Clementina Mesaros, Kevin P. Gillespie, Nathaniel W. Snyder, Robert C. Parry, Andrew J. Worth, Dylan M. Marchione, Ian A. Blair, Carrie A. Sims, and Noelle Saillant
- Subjects
Blood Platelets ,Uncoupling Agents ,General Chemical Engineering ,030204 cardiovascular system & hematology ,Mitochondrion ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Mass Spectrometry ,Cell Line ,Isotopic labeling ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Rotenone ,Humans ,Glycolysis ,Beta oxidation ,General Immunology and Microbiology ,General Neuroscience ,Metabolism ,Mitochondria ,Metabolic pathway ,chemistry ,Biochemistry ,030220 oncology & carcinogenesis ,Isotope Labeling ,Xenobiotic ,Oxidation-Reduction ,Metabolic Networks and Pathways ,Environmental Sciences ,Chromatography, Liquid - Abstract
Perturbed mitochondrial metabolism has received renewed interest as playing a causative role in a range of diseases. Probing alterations to metabolic pathways requires a model in which external factors can be well controlled, allowing for reproducible and meaningful results. Many studies employ transformed cellular models for these purposes; however, metabolic reprogramming that occurs in many cancer cell lines may introduce confounding variables. For this reason primary cells are desirable, though attaining adequate biomass for metabolic studies can be challenging. Here we show that human platelets can be utilized as a platform to carry out metabolic studies in combination with liquid chromatography-tandem mass spectrometry analysis. This approach is amenable to relative quantification and isotopic labeling to probe the activity of specific metabolic pathways. Availability of platelets from individual donors or from blood banks makes this model system applicable to clinical studies and feasible to scale up. Here we utilize isolated platelets to confirm previously identified compensatory metabolic shifts in response to the complex I inhibitor rotenone. More specifically, a decrease in glycolysis is accompanied by an increase in fatty acid oxidation to maintain acetyl-CoA levels. Our results show that platelets can be used as an easily accessible and medically relevant model to probe the effects of xenobiotics on cellular metabolism.
- Published
- 2016
42. Preferential Phosphorylation on Old Histones during Early Mitosis in Human Cells
- Author
-
Yumiao Han, Benjamin A. Garcia, Zuo-Fei Yuan, Shu Lin, and Dylan M. Marchione
- Subjects
0301 basic medicine ,Genomics and Proteomics ,Aurora B kinase ,Mitosis ,Biochemistry ,Methylation ,Histones ,03 medical and health sciences ,Stable isotope labeling by amino acids in cell culture ,Aurora Kinase B ,Humans ,Phosphorylation ,Molecular Biology ,030102 biochemistry & molecular biology ,biology ,Cell Biology ,Cell cycle ,Fibroblasts ,Chromatin ,030104 developmental biology ,Histone ,Acetylation ,biology.protein ,Protein Processing, Post-Translational ,HeLa Cells - Abstract
How histone post-translational modifications (PTMs) are inherited through the cell cycle remains poorly understood. Canonical histones are made in the S phase of the cell cycle. Combining mass spectrometry-based technologies and stable isotope labeling by amino acids in cell culture, we question the distribution of multiple histone PTMs on old versus new histones in synchronized human cells. We show that histone PTMs can be grouped into three categories according to their distributions. Most lysine mono-methylation and acetylation PTMs are either symmetrically distributed on old and new histones or are enriched on new histones. In contrast, most di- and tri-methylation PTMs are enriched on old histones, suggesting that the inheritance of different PTMs is regulated distinctly. Intriguingly, old and new histones are distinct in their phosphorylation status during early mitosis in the following three human cell types: HeLa, 293T, and human foreskin fibroblast cells. The mitotic hallmark H3S10ph is predominantly associated with old H3 at early mitosis and becomes symmetric with the progression of mitosis. This same distribution was observed with other mitotic phosphorylation marks, including H3T3/T6ph, H3.1/2S28ph, and H1.4S26ph but not S28/S31ph on the H3 variant H3.3. Although H3S10ph often associates with the neighboring Lys-9 di- or tri-methylations, they are not required for the asymmetric distribution of Ser-10 phosphorylation on the same H3 tail. Inhibition of the kinase Aurora B does not change the distribution despite significant reduction of H3S10ph levels. However, K9me2 abundance on the new H3 is significantly reduced after Aurora B inhibition, suggesting a cross-talk between H3S10ph and H3K9me2.
- Published
- 2016
43. Abstract A39: Characterizing the epigenetic effects of the histone 3.3 G34W mutation in giant cell tumors of bone
- Author
-
Nada Jabado, Benjamin A. Garcia, Ashot S. Harutyunyan, Dylan M. Marchione, Shriya Deshmukh, and Sima Khazaei
- Subjects
Cancer Research ,Histone ,Oncology ,Histone methyltransferase ,DNA methylation ,Cancer research ,biology.protein ,Epigenetics ,Epigenome ,Biology ,Chromatin immunoprecipitation ,Pediatric cancer ,Epigenomics - Abstract
Introduction: Pediatric glioblastomas (pGBM) are malignant brain tumors associated with a dismal prognosis. A subset of pGBMs carry mutations of either the Lysine 27 or Glycine 34 (G34) amino acid residues of histone 3 variant genes. The same G34 residue is also mutated in 85-95% of giant cell tumors of bone (GCTs), albeit to Tryptophan (G34W) in GCTs rather than to Arginine or Valine (G34R/V) as in pGBMs. The G34 mutation is predicted to impede access of histone methyltransferases like SetD2 to the nearby Lysine 36 residue, thereby altering the epigenome and transcriptome. Methods: To elucidate the tumorigenic effect of G34 mutations, we used the gene-editing technology CRISPR/Cas9 to correct the G34W mutation to wild-type in 2 GCT cell lines. We then investigated CRISPR-edited cell lines using functional assays, proteomic, epigenomic, and transcriptomic analyses. Results: Correction of the G34W mutation to wild-type in CRISPR-edited GCT cells results in phenotypic and functional changes suggestive of reduced tumorigenicity. By mass spectrometry, G34W-mutant GCT cell lines display decreased level of Lysine 36 trimethylation (H3K36me3) on the mutant G34-peptide, similar to G34-mutated pGBM cell lines. However, unlike pGBMs, GCTs display increased levels of Lysine 36 dimethylation (H3K36me2) on the mutant G34W-peptide. Ongoing Experiments and Analyses: We are currently comparing the level and distribution of multiple histone marks by performing chromatin immunoprecipitation followed by next-generation sequencing (ChIP-Seq) of CRISPR-edited wild-type clones relative to the parent G34W-mutant GCT line. We are also assessing differential gene expression by RNA-Seq and characterizing the methylation signature by 850K DNA methylation array of G34W-mutant GCTs and edited clones. Conclusion: The G34W-mutation clearly has an impact on tumorigenic potential, as evidenced by in vitro functional assays. The G34W-mutant peptide of GCT cell lines features a distinct profile of post-translational histone modifications compared to G34R- or G34V-mutant peptides of pGBM cell lines. Further investigation could elucidate the epigenetic mechanism(s) through which the G34W mutation confers its tumorigenic properties. Citation Format: Shriya Deshmukh, Sima Khazaei, Dylan Marchione, Ashot Harutyunyan, Benjamin Garcia, Nada Jabado. Characterizing the epigenetic effects of the histone 3.3 G34W mutation in giant cell tumors of bone [abstract]. In: Proceedings of the AACR Special Conference: Pediatric Cancer Research: From Basic Science to the Clinic; 2017 Dec 3-6; Atlanta, Georgia. Philadelphia (PA): AACR; Cancer Res 2018;78(19 Suppl):Abstract nr A39.
- Published
- 2018
44. Abstract 08: Impaired H3K36 methylation defines a subset of head and neck squamous cell carcinomas
- Author
-
Laurie Ailles, Simon Papillon-Cavanagh, Benjamin A. Garcia, Leonie G. Mikael, David P. Goldstein, Octavia-Maria Dancu, Christina Karamboulas, Peter W. Lewis, Jacek Majewski, John W. Barrett, Denise Bechet, Nada Jabado, Dylan M. Marchione, Sandeep Dhaliwal, Tenzin Gayden, Anthony C. Nichols, William Stecho, Joe S. Mymryk, Ilan Weinreb, Chao Lu, C. David Allis, Jason Karamchandani, and Christopher J. Howlett
- Subjects
Cancer Research ,biology ,Cellular differentiation ,Head and neck cancer ,Cancer ,Epigenome ,Methylation ,medicine.disease ,medicine.disease_cause ,Histone ,Oncology ,DNA methylation ,biology.protein ,medicine ,Cancer research ,Carcinogenesis - Abstract
Human papillomavirus (HPV)-negative head and neck squamous cell carcinomas (HNSCCs) are deadly and common cancers. Recent genomic studies implicate multiple genetic pathways, including cell signaling, cell cycle and immune evasion, in their development. Here we analyze public data sets and uncover a previously unappreciated role of epigenome deregulation in the genesis of 13% of HPV-negative HNSCCs. Specifically, we identify novel recurrent mutations encoding p.Lys36Met (K36M) alterations in multiple H3 histone genes. We further validate the presence of these alterations in multiple independent HNSCC data sets and show that, along with previously described NSD1 mutations, they correspond to a specific DNA methylation cluster. The K36M substitution and NSD1 defects converge on altering methylation of histone H3 at K36 (H3K36), subsequently blocking cellular differentiation and promoting oncogenesis. Our data further indicate limited redundancy for NSD family members in HPV-negative HNSCCs and suggest a potential role for impaired H3K36 methylation in their development. Further investigation of drugs targeting chromatin regulators is warranted in HPV-negative HNSCCs driven by aberrant H3K36 methylation. Citation Format: Chao Lu, Simon Papillon-Cavanagh, Tenzin Gayden, Leonie G. Mikael, Denise Bechet, Christina Karamboulas, Laurie Ailles, Jason Karamchandani, Dylan M. Marchione, Benjamin A. Garcia, Ilan Weinreb, David Goldstein, Peter W. Lewis, Octavia-Maria Dancu, Sandeep Dhaliwal, William Stecho, Christopher J. Howlett, Joe S. Mymryk, John W. Barrett, Anthony C. Nichols, C David Allis, Jacek Majewski, Nada Jabado. Impaired H3K36 methylation defines a subset of head and neck squamous cell carcinomas [abstract]. In: Proceedings of the AACR-AHNS Head and Neck Cancer Conference: Optimizing Survival and Quality of Life through Basic, Clinical, and Translational Research; April 23-25, 2017; San Diego, CA. Philadelphia (PA): AACR; Clin Cancer Res 2017;23(23_Suppl):Abstract nr 08.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.