22 results on '"Joshua P. Plotnik"'
Search Results
2. Figure S1 from Selective Inhibition of the Second Bromodomain of BET Family Proteins Results in Robust Antitumor Activity in Preclinical Models of Acute Myeloid Leukemia
- Author
-
Yu Shen, Marina Konopleva, Warren M. Kati, Keith F. McDaniel, Daniel H. Albert, Michael Boyiadzis, Kathleen A. Dorritie, Neal C. Goodwin, Jenny Rowe, Terrance J. Magoc, Sriram S. Shanmugavelandy, Gaurav Mehta, Debra C. Ferguson, Lina Han, Antonio Cavazos, Qi Zhang, Tamar Uziel, Paul Hessler, Weiguo Feng, Zheng Zha, Xin Lu, Lloyd T. Lam, Vinitha M. Kuruvilla, Emily J. Faivre, Richard J. Bellin, Joshua P. Plotnik, Mai H. Bui, Xiaoli Huang, Xiaoyu Lin, Tianyu Cai, and Lu Zhang
- Abstract
Figure S1.ABBV-744 exhibits potent anti-proliferative activity against AML cells through cell cycle arrest and induction of apoptosis.
- Published
- 2023
- Full Text
- View/download PDF
3. Figure S5 from Selective Inhibition of the Second Bromodomain of BET Family Proteins Results in Robust Antitumor Activity in Preclinical Models of Acute Myeloid Leukemia
- Author
-
Yu Shen, Marina Konopleva, Warren M. Kati, Keith F. McDaniel, Daniel H. Albert, Michael Boyiadzis, Kathleen A. Dorritie, Neal C. Goodwin, Jenny Rowe, Terrance J. Magoc, Sriram S. Shanmugavelandy, Gaurav Mehta, Debra C. Ferguson, Lina Han, Antonio Cavazos, Qi Zhang, Tamar Uziel, Paul Hessler, Weiguo Feng, Zheng Zha, Xin Lu, Lloyd T. Lam, Vinitha M. Kuruvilla, Emily J. Faivre, Richard J. Bellin, Joshua P. Plotnik, Mai H. Bui, Xiaoli Huang, Xiaoyu Lin, Tianyu Cai, and Lu Zhang
- Abstract
Figure S5.Antitumor activity of ABBV-744/venetoclax in AML xenograft model and AML primary samples
- Published
- 2023
- Full Text
- View/download PDF
4. Table S1 from Selective Inhibition of the Second Bromodomain of BET Family Proteins Results in Robust Antitumor Activity in Preclinical Models of Acute Myeloid Leukemia
- Author
-
Yu Shen, Marina Konopleva, Warren M. Kati, Keith F. McDaniel, Daniel H. Albert, Michael Boyiadzis, Kathleen A. Dorritie, Neal C. Goodwin, Jenny Rowe, Terrance J. Magoc, Sriram S. Shanmugavelandy, Gaurav Mehta, Debra C. Ferguson, Lina Han, Antonio Cavazos, Qi Zhang, Tamar Uziel, Paul Hessler, Weiguo Feng, Zheng Zha, Xin Lu, Lloyd T. Lam, Vinitha M. Kuruvilla, Emily J. Faivre, Richard J. Bellin, Joshua P. Plotnik, Mai H. Bui, Xiaoli Huang, Xiaoyu Lin, Tianyu Cai, and Lu Zhang
- Abstract
Primary patient sample information
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Data from Selective Inhibition of the Second Bromodomain of BET Family Proteins Results in Robust Antitumor Activity in Preclinical Models of Acute Myeloid Leukemia
- Author
-
Yu Shen, Marina Konopleva, Warren M. Kati, Keith F. McDaniel, Daniel H. Albert, Michael Boyiadzis, Kathleen A. Dorritie, Neal C. Goodwin, Jenny Rowe, Terrance J. Magoc, Sriram S. Shanmugavelandy, Gaurav Mehta, Debra C. Ferguson, Lina Han, Antonio Cavazos, Qi Zhang, Tamar Uziel, Paul Hessler, Weiguo Feng, Zheng Zha, Xin Lu, Lloyd T. Lam, Vinitha M. Kuruvilla, Emily J. Faivre, Richard J. Bellin, Joshua P. Plotnik, Mai H. Bui, Xiaoli Huang, Xiaoyu Lin, Tianyu Cai, and Lu Zhang
- Abstract
Supplementary figure and table legends
- Published
- 2023
- Full Text
- View/download PDF
6. Table S2 from Selective Inhibition of the Second Bromodomain of BET Family Proteins Results in Robust Antitumor Activity in Preclinical Models of Acute Myeloid Leukemia
- Author
-
Yu Shen, Marina Konopleva, Warren M. Kati, Keith F. McDaniel, Daniel H. Albert, Michael Boyiadzis, Kathleen A. Dorritie, Neal C. Goodwin, Jenny Rowe, Terrance J. Magoc, Sriram S. Shanmugavelandy, Gaurav Mehta, Debra C. Ferguson, Lina Han, Antonio Cavazos, Qi Zhang, Tamar Uziel, Paul Hessler, Weiguo Feng, Zheng Zha, Xin Lu, Lloyd T. Lam, Vinitha M. Kuruvilla, Emily J. Faivre, Richard J. Bellin, Joshua P. Plotnik, Mai H. Bui, Xiaoli Huang, Xiaoyu Lin, Tianyu Cai, and Lu Zhang
- Abstract
Tumor growth inhibition in cell line-based xenograft models and supporting statistics.
- Published
- 2023
- Full Text
- View/download PDF
7. Data from Selective Inhibition of the Second Bromodomain of BET Family Proteins Results in Robust Antitumor Activity in Preclinical Models of Acute Myeloid Leukemia
- Author
-
Yu Shen, Marina Konopleva, Warren M. Kati, Keith F. McDaniel, Daniel H. Albert, Michael Boyiadzis, Kathleen A. Dorritie, Neal C. Goodwin, Jenny Rowe, Terrance J. Magoc, Sriram S. Shanmugavelandy, Gaurav Mehta, Debra C. Ferguson, Lina Han, Antonio Cavazos, Qi Zhang, Tamar Uziel, Paul Hessler, Weiguo Feng, Zheng Zha, Xin Lu, Lloyd T. Lam, Vinitha M. Kuruvilla, Emily J. Faivre, Richard J. Bellin, Joshua P. Plotnik, Mai H. Bui, Xiaoli Huang, Xiaoyu Lin, Tianyu Cai, and Lu Zhang
- Abstract
Dual bromodomain BET inhibitors that bind with similar affinities to the first and second bromodomains across BRD2, BRD3, BRD4, and BRDT have displayed modest activity as monotherapy in clinical trials. Thrombocytopenia, closely followed by symptoms characteristic of gastrointestinal toxicity, have presented as dose-limiting adverse events that may have prevented escalation to higher dose levels required for more robust efficacy. ABBV-744 is a highly selective inhibitor for the second bromodomain of the four BET family proteins. In contrast to the broad antiproliferative activities observed with dual bromodomain BET inhibitors, ABBV-744 displayed significant antiproliferative activities largely although not exclusively in cancer cell lines derived from acute myeloid leukemia and androgen receptor positive prostate cancer. Studies in acute myeloid leukemia xenograft models demonstrated antitumor efficacy for ABBV-744 that was comparable with the pan-BET inhibitor ABBV-075 but with an improved therapeutic index. Enhanced antitumor efficacy was also observed with the combination of ABBV-744 and the BCL-2 inhibitor, venetoclax compared with monotherapies of either agent alone. These results collectively support the clinical evaluation of ABBV-744 in AML (Clinical Trials.gov identifier: NCT03360006).
- Published
- 2023
- Full Text
- View/download PDF
8. Figure S3 from Selective Inhibition of the Second Bromodomain of BET Family Proteins Results in Robust Antitumor Activity in Preclinical Models of Acute Myeloid Leukemia
- Author
-
Yu Shen, Marina Konopleva, Warren M. Kati, Keith F. McDaniel, Daniel H. Albert, Michael Boyiadzis, Kathleen A. Dorritie, Neal C. Goodwin, Jenny Rowe, Terrance J. Magoc, Sriram S. Shanmugavelandy, Gaurav Mehta, Debra C. Ferguson, Lina Han, Antonio Cavazos, Qi Zhang, Tamar Uziel, Paul Hessler, Weiguo Feng, Zheng Zha, Xin Lu, Lloyd T. Lam, Vinitha M. Kuruvilla, Emily J. Faivre, Richard J. Bellin, Joshua P. Plotnik, Mai H. Bui, Xiaoli Huang, Xiaoyu Lin, Tianyu Cai, and Lu Zhang
- Abstract
Figure S3.Comparison of ABBV-744 and GSK046 BD2 selective compounds.
- Published
- 2023
- Full Text
- View/download PDF
9. Figure S6 from Selective Inhibition of the Second Bromodomain of BET Family Proteins Results in Robust Antitumor Activity in Preclinical Models of Acute Myeloid Leukemia
- Author
-
Yu Shen, Marina Konopleva, Warren M. Kati, Keith F. McDaniel, Daniel H. Albert, Michael Boyiadzis, Kathleen A. Dorritie, Neal C. Goodwin, Jenny Rowe, Terrance J. Magoc, Sriram S. Shanmugavelandy, Gaurav Mehta, Debra C. Ferguson, Lina Han, Antonio Cavazos, Qi Zhang, Tamar Uziel, Paul Hessler, Weiguo Feng, Zheng Zha, Xin Lu, Lloyd T. Lam, Vinitha M. Kuruvilla, Emily J. Faivre, Richard J. Bellin, Joshua P. Plotnik, Mai H. Bui, Xiaoli Huang, Xiaoyu Lin, Tianyu Cai, and Lu Zhang
- Abstract
Figure S6.Identification of biomarker of response to ABBV-744/venetoclax therapy.
- Published
- 2023
- Full Text
- View/download PDF
10. Figure S4 from Selective Inhibition of the Second Bromodomain of BET Family Proteins Results in Robust Antitumor Activity in Preclinical Models of Acute Myeloid Leukemia
- Author
-
Yu Shen, Marina Konopleva, Warren M. Kati, Keith F. McDaniel, Daniel H. Albert, Michael Boyiadzis, Kathleen A. Dorritie, Neal C. Goodwin, Jenny Rowe, Terrance J. Magoc, Sriram S. Shanmugavelandy, Gaurav Mehta, Debra C. Ferguson, Lina Han, Antonio Cavazos, Qi Zhang, Tamar Uziel, Paul Hessler, Weiguo Feng, Zheng Zha, Xin Lu, Lloyd T. Lam, Vinitha M. Kuruvilla, Emily J. Faivre, Richard J. Bellin, Joshua P. Plotnik, Mai H. Bui, Xiaoli Huang, Xiaoyu Lin, Tianyu Cai, and Lu Zhang
- Abstract
Figure S4.ABBV-744 displaces BRD4 from regulatory regions of BCL2, BCL2L1 and RUNX1 in sensitive AML cells but not insensitive AML line.
- Published
- 2023
- Full Text
- View/download PDF
11. Abstract 6156: MALT1 protease inhibition overcomes BTK inhibitor resistance and shows synergistic activity with venetoclax in models of B cell lymphoma and leukemia
- Author
-
Joshua P. Plotnik, Raghuveer S. Mali, Velitchka Bontcheva, Colleen Dowell, Jun Chen, Adam E. Richardson, Ryan A. McClure, Paul Jung, Lan Pham, Andrew J. Souers, Jonathan A. Meulbroek, and William N. Pappano
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Specific B cell malignancies, including CLL and the aggressive non-GCB subtype of DLBCL, are driven by constitutive activation of the B cell receptor (BCR) pathway and the transcription factor NF-κB. Pharmacological inhibition of MALT1 protease, a key mediator of the BCR/NF-κB signal transduction pathway, may therefore provide an attractive treatment option for patients with these cancers. Further, as combination therapy is often required for the treatment of aggressive B cell malignancies, the identification of therapies that synergistically combine with MALT1 inhibitors could afford additional and promising treatment options. Experimental Procedures: A highly potent and orally bioavailable MALT1 protease inhibitor (ABBV-MALT1) was used to test the hypothesis that MALT1 inhibition will abrogate the proliferation of preclinical models of B cell malignancies in vitro and in vivo. Tumors treated with ABBV-MALT1 were subjected to transcriptomic and functional proteomic assays to elucidate molecular mechanisms of action and rational combination partners. Results: Mechanistic studies reveal that ABBV-MALT1 effectively inhibits signal transduction of the BCR pathway and reduces NF-κB gene activation in non-GCB DLBCL cell lines resulting in cell cycle arrest and diminished viability. In vivo, oral administration of this compound demonstrates robust tumor growth inhibition in several models of B cell tumors, including non-GCB DLBCL models that are resistant to Bruton’s tyrosine kinase (BTK) inhibitors. NF-κB target genes include the pro-survival family members BCL-XL and BCL2-A1, which aid in regulation of the intrinsic apoptosis pathway. As ABBV-MALT1-induced inhibition of the NF-κB pathway resulted in downregulation of these genes, we hypothesized that the associated tumor models would become increasingly dependent on the pro-survival family member BCL-2. To test this hypothesis, combination studies of ABBV-MALT1 and the selective BCL-2 inhibitor venetoclax were performed in both cell line and patient-derived xenograft models of DLBCL. Herein we show that concomitant administration of ABBV-MALT1 and venetoclax results in dramatic antitumor activity in all models tested in vivo. This efficacy also translates to primary patient CLL cells in vitro where the combination confers greater levels of apoptosis compared to either agent alone. Conclusion: ABBV-MALT1 demonstrates robust single agent anti-tumor activity in malignant B cell models that are resistant to BTK inhibitors. Moreover, combination of ABBV-MALT1 with the BCL-2 inhibitor venetoclax shows synergistic cell killing of B cell tumors in vitro and dramatic tumor regression in vivo. Together, these data indicate that MALT1 inhibition may overcome BTK inhibitor resistance and combine with venetoclax to effectively treat patients with B cell malignancies. Citation Format: Joshua P. Plotnik, Raghuveer S. Mali, Velitchka Bontcheva, Colleen Dowell, Jun Chen, Adam E. Richardson, Ryan A. McClure, Paul Jung, Lan Pham, Andrew J. Souers, Jonathan A. Meulbroek, William N. Pappano. MALT1 protease inhibition overcomes BTK inhibitor resistance and shows synergistic activity with venetoclax in models of B cell lymphoma and leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6156.
- Published
- 2023
- Full Text
- View/download PDF
12. Selective inhibition of the BD2 bromodomain of BET proteins in prostate cancer
- Author
-
Denise Wilcox, Vasudha Sehgal, Daniel H. Albert, John K. Pratt, Keith F. McDaniel, Xin Lu, Chaohong Sun, Dachun Liu, Joshua P. Plotnik, Srinivasa R. Mantena, Emily J. Faivre, Lisa A. Hasvold, George S. Sheppard, Xiaoli Huang, Le Wang, Lance J Bigelow, Stacey Fossey, Steve D. Fidanze, Lloyd T. Lam, Chang H. Park, Sanjay C. Panchal, Warren M. Kati, John J. Nicolette, Richard J. Bellin, Gaurav Mehta, Xiaoyu Lin, Mai H. Bui, Lu Zhang, Paul Hessler, Maricel Torrent, Tamar Uziel, Saul H. Rosenberg, Yu Shen, and Kenton L. Longenecker
- Subjects
Male ,BRD4 ,Transcription, Genetic ,Pyridines ,Cell Cycle Proteins ,BET inhibitor ,Mice ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Protein Domains ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Gene silencing ,Pyrroles ,Receptor ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,biology ,Chemistry ,Prostatic Neoplasms ,medicine.disease ,Xenograft Model Antitumor Assays ,Rats ,Bromodomain ,Gene Expression Regulation, Neoplastic ,Enhancer Elements, Genetic ,Histone ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Chromatin immunoprecipitation ,Transcription Factors - Abstract
Proteins of the bromodomain and extra-terminal (BET) domain family are epigenetic readers that bind acetylated histones through their bromodomains to regulate gene transcription. Dual-bromodomain BET inhibitors (DbBi) that bind with similar affinities to the first (BD1) and second (BD2) bromodomains of BRD2, BRD3, BRD4 and BRDt have displayed modest clinical activity in monotherapy cancer trials. A reduced number of thrombocytes in the blood (thrombocytopenia) as well as symptoms of gastrointestinal toxicity are dose-limiting adverse events for some types of DbBi1–5. Given that similar haematological and gastrointestinal defects were observed after genetic silencing of Brd4 in mice6, the platelet and gastrointestinal toxicities may represent on-target activities associated with BET inhibition. The two individual bromodomains in BET family proteins may have distinct functions7–9 and different cellular phenotypes after pharmacological inhibition of one or both bromodomains have been reported10,11, suggesting that selectively targeting one of the bromodomains may result in a different efficacy and tolerability profile compared with DbBi. Available compounds that are selective to individual domains lack sufficient potency and the pharmacokinetics properties that are required for in vivo efficacy and tolerability assessment10–13. Here we carried out a medicinal chemistry campaign that led to the discovery of ABBV-744, a highly potent and selective inhibitor of the BD2 domain of BET family proteins with drug-like properties. In contrast to the broad range of cell growth inhibition induced by DbBi, the antiproliferative activity of ABBV-744 was largely, but not exclusively, restricted to cell lines of acute myeloid leukaemia and prostate cancer that expressed the full-length androgen receptor (AR). ABBV-744 retained robust activity in prostate cancer xenografts, and showed fewer platelet and gastrointestinal toxicities than the DbBi ABBV-07514. Analyses of RNA expression and chromatin immunoprecipitation followed by sequencing revealed that ABBV-744 displaced BRD4 from AR-containing super-enhancers and inhibited AR-dependent transcription, with less impact on global transcription compared with ABBV-075. These results underscore the potential value of selectively targeting the BD2 domain of BET family proteins for cancer therapy. ABBV-744, a selective inhibitor of the BD2 domains of BET family proteins, is effective against prostate cancer in mouse xenograft models, with lower toxicities than the dual-bromodomain BET inhibitor ABBV-075.
- Published
- 2020
- Full Text
- View/download PDF
13. Selective Inhibition of the Second Bromodomain of BET Family Proteins Results in Robust Antitumor Activity in Preclinical Models of Acute Myeloid Leukemia
- Author
-
Marina Konopleva, Tamar Uziel, Xiaoli Huang, Weiguo Feng, Xiaoyu Lin, Warren M. Kati, Lloyd T. Lam, Vinitha Mary Kuruvilla, Mai H. Bui, Emily J. Faivre, Tianyu Cai, Jenny Rowe, Daniel H. Albert, Richard J. Bellin, Lu Zhang, Zheng Zha, Sriram S. Shanmugavelandy, Paul Hessler, Michael Boyiadzis, Gaurav Mehta, Antonio Cavazos, Keith F. McDaniel, Joshua P. Plotnik, Terrance J. Magoc, Xin Lu, Debra Ferguson, Yu Shen, Lina Han, Neal Goodwin, Qi Zhang, and Kathleen A. Dorritie
- Subjects
Cancer Research ,BRD4 ,Pyridines ,Androgen Receptor Positive ,Antineoplastic Agents ,Apoptosis ,Mice, SCID ,BET inhibitor ,Prostate cancer ,chemistry.chemical_compound ,Mice ,Therapeutic index ,Mice, Inbred NOD ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,Pyrroles ,Cell Proliferation ,Sulfonamides ,Venetoclax ,business.industry ,Myeloid leukemia ,Proteins ,medicine.disease ,Bridged Bicyclo Compounds, Heterocyclic ,Xenograft Model Antitumor Assays ,Bromodomain ,Leukemia, Myeloid, Acute ,Oncology ,chemistry ,Proto-Oncogene Proteins c-bcl-2 ,Cancer research ,Drug Therapy, Combination ,Female ,business - Abstract
Dual bromodomain BET inhibitors that bind with similar affinities to the first and second bromodomains across BRD2, BRD3, BRD4, and BRDT have displayed modest activity as monotherapy in clinical trials. Thrombocytopenia, closely followed by symptoms characteristic of gastrointestinal toxicity, have presented as dose-limiting adverse events that may have prevented escalation to higher dose levels required for more robust efficacy. ABBV-744 is a highly selective inhibitor for the second bromodomain of the four BET family proteins. In contrast to the broad antiproliferative activities observed with dual bromodomain BET inhibitors, ABBV-744 displayed significant antiproliferative activities largely although not exclusively in cancer cell lines derived from acute myeloid leukemia and androgen receptor positive prostate cancer. Studies in acute myeloid leukemia xenograft models demonstrated antitumor efficacy for ABBV-744 that was comparable with the pan-BET inhibitor ABBV-075 but with an improved therapeutic index. Enhanced antitumor efficacy was also observed with the combination of ABBV-744 and the BCL-2 inhibitor, venetoclax compared with monotherapies of either agent alone. These results collectively support the clinical evaluation of ABBV-744 in AML (Clinical Trials.gov identifier: NCT03360006).
- Published
- 2021
14. ETS1 induction by the microenvironment promotes ovarian cancer metastasis through focal adhesion kinase
- Author
-
Robert E. Emerson, Peter C. Hollenhorst, Sunil Tomar, Joshua Scantland, Subramanyam Dasari, Zahir Sheikh, Joshua P. Plotnik, James Haley, Dean Lenz, and Anirban K. Mitra
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Transplantation, Heterologous ,Mice, Nude ,Kaplan-Meier Estimate ,Biology ,Metastasis ,Proto-Oncogene Protein c-ets-1 ,Focal adhesion ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,ETS1 ,Cell Movement ,Cell Line, Tumor ,Internal medicine ,Tumor Microenvironment ,medicine ,Transcriptional regulation ,Animals ,Humans ,Neoplasm Metastasis ,Transcription factor ,Ovarian Neoplasms ,Tumor microenvironment ,medicine.disease ,Juxtacrine signalling ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Focal Adhesion Kinase 1 ,030220 oncology & carcinogenesis ,Cancer research ,Female ,RNA Interference - Abstract
Metastatic colonization involves paracrine/juxtacrine interactions with the microenvironment inducing an adaptive response through transcriptional regulation. However, the identities of transcription factors (TFs) induced by the metastatic microenvironment in ovarian cancer (OC) and their mechanism of action is poorly understood. Using an organotypic 3D culture model recapitulating the early events of metastasis, we identified ETS1 as the most upregulated member of the ETS family of TFs in metastasizing OC cells as they interacted with the microenvironment. ETS1 was regulated by p44/42 MAP kinase signaling activated in the OC cells interacting with mesothelial cells at the metastatic site. Human OC tumors had increased expression of ETS1, which predicted poor prognosis. ETS1 regulated OC metastasis both in vitro and in mouse xenografts. A combination of ChIP-seq and RNA-seq analysis and functional rescue experiments revealed FAK as the key transcriptional target and downstream effector of ETS1. Taken together, our results indicate that ETS1 is an essential transcription factor induced in OC cells by the microenvironment, which promotes metastatic colonization though the transcriptional upregulation of its target FAK.
- Published
- 2018
- Full Text
- View/download PDF
15. Fighting PRC1 via the RING
- Author
-
Joshua P. Plotnik
- Subjects
0303 health sciences ,Chemistry ,fungi ,030302 biochemistry & molecular biology ,Repressor ,macromolecular substances ,Cell Biology ,Computational biology ,Ring (chemistry) ,Bench to bedside ,Chromatin ,03 medical and health sciences ,Posttranslational modification ,PRC1 ,Molecular Biology ,030304 developmental biology - Abstract
Over the past two decades, inhibitors of the polycomb repressor complex (PRC) have been driven from bench to bedside. New chemical compounds targeting the RING1B in polycomb repressor complex 1 (PRC1) add important tools to regulate polycomb functions.
- Published
- 2021
- Full Text
- View/download PDF
16. An Interaction with Ewing’s Sarcoma Breakpoint Protein EWS Defines a Specific Oncogenic Mechanism of ETS Factors Rearranged in Prostate Cancer
- Author
-
Nagarathinam Selvaraj, Vivekananda Kedage, Justin A. Budka, Taylor R. Nicholas, Joshua P. Plotnik, Travis J. Jerde, and Peter C. Hollenhorst
- Subjects
Male ,0301 basic medicine ,Carcinogenesis ,Mice, Nude ,Sarcoma, Ewing ,Chromosomal rearrangement ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,ETV1 ,Proto-Oncogene Protein c-ets-1 ,03 medical and health sciences ,Prostate cancer ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein Interaction Domains and Motifs ,EWS ,Promoter Regions, Genetic ,lcsh:QH301-705.5 ,Transcription factor ,Cell Proliferation ,Gene Rearrangement ,Regulation of gene expression ,Genetics ,Breakpoint ,Prostatic Neoplasms ,Ewing's sarcoma ,Oncogenes ,prostate cancer ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Phenotype ,030104 developmental biology ,lcsh:Biology (General) ,ERG ,Sarcoma ,Ewing’s sarcoma ,RNA-Binding Protein EWS ,ETS ,Protein Binding ,Transcription Factors - Abstract
Summary: More than 50% of prostate tumors have a chromosomal rearrangement resulting in aberrant expression of an oncogenic ETS family transcription factor. However, mechanisms that differentiate the function of oncogenic ETS factors expressed in prostate tumors from non-oncogenic ETS factors expressed in normal prostate are unknown. Here, we find that four oncogenic ETS (ERG, ETV1, ETV4, and ETV5), and no other ETS, interact with the Ewing’s sarcoma breakpoint protein, EWS. This EWS interaction was necessary and sufficient for oncogenic ETS functions including gene activation, cell migration, clonogenic survival, and transformation. Significantly, the EWS interacting region of ERG has no homology with that of ETV1, ETV4, and ETV5. Therefore, this finding may explain how divergent ETS factors have a common oncogenic function. Strikingly, EWS is fused to various ETS factors by the chromosome translocations that cause Ewing’s sarcoma. Therefore, these findings link oncogenic ETS function in both prostate cancer and Ewing’s sarcoma. : A subset of ETS transcription factors is oncogenic in prostate. Kedage et al. show that oncogenic ETS, but not other ETS, interact with EWS, and this interaction is necessary for oncogenic functions. Because EWS is fused to ETS factors in Ewing’s sarcoma, this finding links the mechanisms of these diseases. Keywords: prostate cancer, ETS, EWS, Ewing’s sarcoma, ERG
- Published
- 2016
- Full Text
- View/download PDF
17. Extracellular Signal-Regulated Kinase Signaling Regulates the Opposing Roles of JUN Family Transcription Factors at ETS/AP-1 Sites and in Cell Migration
- Author
-
Peter C. Hollenhorst, Nagarathinam Selvaraj, Justin A. Budka, Joshua P. Plotnik, and Mary W. Ferris
- Subjects
Male ,MAPK/ERK pathway ,Proto-Oncogene Proteins c-jun ,JUNB ,Biology ,Cell Movement ,Cell Line, Tumor ,Humans ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Molecular Biology ,Transcription factor ,Regulation of gene expression ,Sequence Analysis, RNA ,Kinase ,HEK 293 cells ,Prostatic Neoplasms ,Cell migration ,Articles ,Cell Biology ,Gene Expression Regulation, Neoplastic ,Transcription Factor AP-1 ,HEK293 Cells ,ras Proteins ,Cancer research ,Signal transduction ,K562 Cells ,Protein Binding ,Signal Transduction ,Transcription Factors - Abstract
JUN transcription factors bind DNA as part of the AP-1 complex, regulate many cellular processes, and play a key role in oncogenesis. The three JUN proteins (c-JUN, JUNB, and JUND) can have both redundant and unique functions depending on the biological phenotype and cell type assayed. Mechanisms that allow this dynamic switching between overlapping and distinct functions are unclear. Here we demonstrate that JUND has a role in prostate cell migration that is the opposite of c-JUN's and JUNB's. RNA sequencing reveals that opposing regulation by c-JUN and JUND defines a subset of AP-1 target genes with cell migration roles. cis-regulatory elements for only this subset of targets were enriched for ETS factor binding, indicating a specificity mechanism. Interestingly, the function of c-JUN and JUND in prostate cell migration switched when we compared cells with an inactive versus an active RAS/extracellular signal-regulated kinase (ERK) signaling pathway. We show that this switch is due to phosphorylation and activation of JUND by ERK. Thus, the ETS/AP-1 sequence defines a unique gene expression program regulated by the relative levels of JUN proteins and RAS/ERK signaling. This work provides a rationale for how transcription factors can have distinct roles depending on the signaling status and the biological function in question.
- Published
- 2015
- Full Text
- View/download PDF
18. ETS1 is a genome-wide effector of RAS/ERK signaling in epithelial cells
- Author
-
Justin A. Budka, Mary W. Ferris, Joshua P. Plotnik, and Peter C. Hollenhorst
- Subjects
Transcriptional Activation ,MAPK/ERK pathway ,MAP Kinase Signaling System ,Biology ,Proto-Oncogene Protein c-ets-1 ,Proto-Oncogene Proteins p21(ras) ,ETS1 ,Cell Movement ,Cell Line, Tumor ,Anti-apoptotic Ras signalling cascade ,Genetics ,Humans ,Regulatory Elements, Transcriptional ,Extracellular Signal-Regulated MAP Kinases ,Cells, Cultured ,Binding Sites ,Proto-Oncogene Proteins c-ets ,Extracellular matrix-cell signaling ,Genome, Human ,Gene regulation, Chromatin and Epigenetics ,Carcinoma ,Epithelial Cells ,Cell migration ,Chromatin ,Transcription Factor AP-1 ,Cancer research ,Caco-2 Cells ,Signal transduction - Abstract
The RAS/ERK pathway is commonly activated in carcinomas and promotes oncogenesis by altering transcriptional programs. However, the array of cis-regulatory elements and trans-acting factors that mediate these transcriptional changes is still unclear. Our genome-wide analysis determined that a sequence consisting of neighboring ETS and AP-1 transcription factor binding sites is enriched near cell migration genes activated by RAS/ERK signaling in epithelial cells. In vivo screening of candidate ETS proteins revealed that ETS1 is specifically required for migration of RAS/ERK activated cells. Furthermore, both migration and transcriptional activation through ETS/AP-1 required ERK phosphorylation of ETS1. Genome-wide mapping of multiple ETS proteins demonstrated that ETS1 binds specifically to enhancer ETS/AP-1 sequences. ETS1 occupancy, and its role in cell migration, was conserved in epithelial cells derived from multiple tissues, consistent with a chromatin organization common to epithelial cell lines. Genome-wide expression analysis showed that ETS1 was required for activation of RAS-regulated cell migration genes, but also identified a surprising role for ETS1 in the repression of genes such as DUSP4, DUSP6 and SPRY4 that provide negative feedback to the RAS/ERK pathway. Consistently, ETS1 was required for robust RAS/ERK pathway activation. Therefore, ETS1 has dual roles in mediating epithelial-specific RAS/ERK transcriptional functions.
- Published
- 2014
- Full Text
- View/download PDF
19. Genome-Wide Analysis of RAS/ERK Signaling Targets
- Author
-
Joshua P, Plotnik and Peter C, Hollenhorst
- Subjects
Gene Expression Profiling ,ras Proteins ,Computational Biology ,High-Throughput Nucleotide Sequencing ,Humans ,Extracellular Signal-Regulated MAP Kinases ,Transcriptome ,Genome-Wide Association Study ,Signal Transduction - Abstract
Identifying gene expression changes mediated by signaling pathways is necessary to determine mechanisms that cause phenotypic change. Recent advances in next-generation sequencing and informatic pipelines have streamlined the ability for laboratories to create and analyze transcriptomic data. Here, we describe the preparation of samples and transcriptomic analysis in order to determine gene expression programs regulated by RAS/ERK signaling.
- Published
- 2016
20. Genome-Wide Analysis of RAS/ERK Signaling Targets
- Author
-
Joshua P. Plotnik and Peter C. Hollenhorst
- Subjects
0301 basic medicine ,Genetics ,RNA-Seq ,Genome-wide association study ,Computational biology ,Biology ,Phenotype ,Gene expression profiling ,Transcriptome ,03 medical and health sciences ,030104 developmental biology ,Anti-apoptotic Ras signalling cascade ,Gene expression ,sense organs ,Signal transduction - Abstract
Identifying gene expression changes mediated by signaling pathways is necessary to determine mechanisms that cause phenotypic change. Recent advances in next-generation sequencing and informatic pipelines have streamlined the ability for laboratories to create and analyze transcriptomic data. Here, we describe the preparation of samples and transcriptomic analysis in order to determine gene expression programs regulated by RAS/ERK signaling.
- Published
- 2016
- Full Text
- View/download PDF
21. Interaction with ZMYND11 mediates opposing roles of Ras-responsive transcription factors ETS1 and ETS2
- Author
-
Joshua P. Plotnik and Peter C. Hollenhorst
- Subjects
0301 basic medicine ,Male ,Lung Neoplasms ,Transcription, Genetic ,Cell Cycle Proteins ,Biology ,Adenocarcinoma ,medicine.disease_cause ,Proto-Oncogene Protein c-ets-2 ,Proto-Oncogene Protein c-ets-1 ,03 medical and health sciences ,0302 clinical medicine ,ETS1 ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Transcriptional attenuation ,Transcription factor ,Regulation of gene expression ,Oncogene ,HEK 293 cells ,Gene regulation, Chromatin and Epigenetics ,Prostatic Neoplasms ,Survival Analysis ,Cell biology ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,HEK293 Cells ,A549 Cells ,Organ Specificity ,030220 oncology & carcinogenesis ,ras Proteins ,Signal transduction ,Mitogen-Activated Protein Kinases ,Carcinogenesis ,Carrier Proteins ,Co-Repressor Proteins ,Protein Binding ,Signal Transduction - Abstract
Aberrant activation of RAS/MAPK signaling is a driver of over one third of all human carcinomas. The homologous transcription factors ETS1 and ETS2 mediate activation of gene expression programs downstream of RAS/MAPK signaling. ETS1 is important for oncogenesis in many tumor types. However, ETS2 can act as an oncogene in some cellular backgrounds, and as a tumor suppressor in others, and the molecular mechanism responsible for this cell-type specific function remains unknown. Here, we show that ETS1 and ETS2 can regulate a cell migration gene expression program in opposite directions, and provide the first comparison of the ETS1 and ETS2 cistromes. This genomic data and an ETS1 deletion line reveal that the opposite function of ETS2 is a result of binding site competition and transcriptional attenuation due to weaker transcriptional activation by ETS2 compared to ETS1. This weaker activation was mapped to the ETS2 N-terminus and a specific interaction with the co-repressor ZMYND11 (BS69). Furthermore, ZMYND11 expression levels in patient tumors correlated with oncogenic versus tumor suppressive roles of ETS2. Therefore, these data indicate a novel and specific mechanism allowing ETS2 to switch between oncogenic and tumor suppressive functions in a cell-type specific manner.
- Published
- 2016
22. Abstract A34: Induction of a novel ETS1/FAK pathway in metastasizing ovarian cancer cells by the omental microenvironment primes them for metastatic colonization
- Author
-
Joshua P. Plotnik, Zahir Sheikh, Robert E. Emerson, Anirban K. Mitra, Joshua Scantland, James Haley, Dean Lenz, Sunil Tomar, and Peter C. Hollenhorst
- Subjects
Cancer Research ,Tissue microarray ,Cancer ,Biology ,medicine.disease ,Metastasis ,Paracrine signalling ,Oncology ,Downregulation and upregulation ,ETS1 ,Cancer cell ,Cancer research ,medicine ,Ovarian cancer - Abstract
Metastatic colonization of ovarian cancer involves productive paracrine/juxtacrine interactions with the microenvironment. The resulting induction of an adaptive response in the cancer cells enables them to establish themselves in the new microenvironment and take advantage of the new factors available. A key feature of this adaptation is induced changes in gene expression through transcriptional regulation as a result of microenvironmental cues. However, the identities of transcription factors induced by the metastatic microenvironment in ovarian cancer and their mechanism of action are poorly understood. Using an organotypic 3D culture model recapitulating the early events of metastasis, we identified ETS1, a member of the ETS family of TFs, as an essential driver of metastatic colonization. Increased ETS1 expression was induced in metastasizing ovarian cancer cells interacting with the mesothelial cells covering the surface of the omentum. The mechanism of upregulation was through the activation of p44/42 MAP kinase signaling in the cancer cells induced by TGFbeta from the microenvironment. We also found an increased ETS1 expression in human ovarian cancer samples as compared to normal fallopian tubes using a tissue microarray. Moreover, higher expression of ETS1 was a predictor of poor prognosis in ovarian cancer patients. Knocking down ETS1 decreased migration, proliferation, and colony formation as well as invasion through and colonization of the organotypic 3D culture. Overexpression of ETS1 had the opposite effect. CRISPR/Cas9-mediated knockout of ETS1 resulted in decreased tumor burden in mouse xenografts. A combination of ChIP-seq and RNA-seq analysis revealed that ETS1 promoted an EMT phenotype and FAK was identified as a novel transcriptional target. Inhibition of FAK functionally mimicked the effects of ETS1 inhibition in the ovarian cancer cells. Moreover, functional rescue experiments established FAK as a downstream effector of ETS1 during ovarian cancer metastasis. Taken together, our results indicate that ETS1 is an essential transcription factor induced in ovarian cancer cells by the microenvironment, which promotes metastatic colonization. This is the first report establishing FAK as a transcriptional target and functional effector of ETS1 in establishing metastatic tumors. Citation Format: Sunil Tomar, Joshua P. Plotnik, James Haley, Joshua Scantland, Zahir Sheikh, Robert Emerson, Dean Lenz, Peter C. Hollenhorst, Anirban K. Mitra. Induction of a novel ETS1/FAK pathway in metastasizing ovarian cancer cells by the omental microenvironment primes them for metastatic colonization. [abstract]. In: Proceedings of the AACR Conference: Addressing Critical Questions in Ovarian Cancer Research and Treatment; Oct 1-4, 2017; Pittsburgh, PA. Philadelphia (PA): AACR; Clin Cancer Res 2018;24(15_Suppl):Abstract nr A34.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.