25 results on '"Justin A. Budka"'
Search Results
2. Supplementary Fig. 1 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Specificity of TFE3 antibody across TFE3 oncoproteins
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Table S7. from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
A complete list of targeted genes in the PI3K/AKT signaling pathway with their associated miRNA from cluster 3.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Table S3. from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Comprehensive panels of 287 KEGG pathways associated identified by bioinformatics analysis of TFE3 ChIP-seq peaks in RP-R07 cells.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Table S4. from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Comprehensive panels of 96 PANTHER pathways identified by bioinformatics analysis of TFE3 ChIP-seq peaks in RP-R07 cells.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Table S1 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Quality control for ChIP-seq
- Published
- 2023
- Full Text
- View/download PDF
7. Data from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Purpose:Translocation renal cell carcinoma (tRCC) represents a rare subtype of kidney cancer associated with various TFE3, TFEB, or MITF gene fusions that are not responsive to standard treatments for RCC. Therefore, the identification of new therapeutic targets represents an unmet need for this disease.Experimental Design:We have established and characterized a tRCC patient-derived xenograft, RP-R07, as a novel preclinical model for drug development by using next-generation sequencing and bioinformatics analysis. We then assessed the therapeutic potential of inhibiting the identified pathway using in vitro and in vivo models.Results:The presence of a SFPQ-TFE3 fusion [t(X;1) (p11.2; p34)] with chromosomal break-points was identified by RNA-seq and validated by RT-PCR. TFE3 chromatin immunoprecipitation followed by deep sequencing analysis indicated a strong enrichment for the PI3K/AKT/mTOR pathway. Consistently, miRNA microarray analysis also identified PI3K/AKT/mTOR as a highly enriched pathway in RP-R07. Upregulation of PI3/AKT/mTOR pathway in additional TFE3–tRCC models was confirmed by significantly higher expression of phospho-S6 (P < 0.0001) and phospho-4EBP1 (P < 0.0001) in established tRCC cell lines compared with clear cell RCC cells. Simultaneous vertical targeting of both PI3K/AKT and mTOR axis provided a greater antiproliferative effect both in vitro (P < 0.0001) and in vivo (P < 0.01) compared with single-node inhibition. Knockdown of TFE3 in RP-R07 resulted in decreased expression of IRS-1 and inhibited cell proliferation.Conclusions:These results identify TFE3/IRS-1/PI3K/AKT/mTOR as a potential dysregulated pathway in TFE3–tRCC, and suggest a therapeutic potential of vertical inhibition of this axis by using a dual PI3K/mTOR inhibitor for patients with TFE3–tRCC.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Fig. 3 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Pie chart for TFE3 binding sites
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Table S6. from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Lists of statistically enriched pathways targeted by differential expression of miRNA in cluster 3.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Fig. 6 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Effect of different rapamycin concentrations
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Fig. 5 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Enriched KEGG PI3K/AKT signaling pathway visualization
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Fig. 4 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Pie chart for TFE3 binding sites
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Table S5. from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Comprehensive panels of 403 WIKI tools pathways identified by bioinformatics analysis of TFE3 ChIP-seq peaks in RP-R07 cells.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Fig. 2 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
ChiP-seq for TFE3 in UOK-146
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Fig. 8 from Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Roberto Pili, Peter C. Hollenhorst, George M. Yousef, W. Marston Linehan, Chinghai Kao, Sreenivasulu Chintala, Ashley Orillion, May Elbanna, Remi Adelaiye-Ogala, Venkata Nithinsai Chintala, Khunsha Ahmed, Anthony C. Wood, Eric Kauffman, Sheng Yu Ku, Mary W. Ferris, Heba W.Z Khella, Justin A. Budka, and Nur P. Damayanti
- Abstract
Schema for multimodal inhibition in tRCC
- Published
- 2023
- Full Text
- View/download PDF
16. Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma
- Author
-
Venkata Nithinsai Chintala, George M. Yousef, Sreenivasulu Chintala, Anthony C. Wood, Nur P. Damayanti, Remi Adelaiye-Ogala, Chinghai Kao, Sheng-Yu Ku, Mary W. Ferris, Roberto Pili, Peter C. Hollenhorst, May Elbanna, Justin A. Budka, Eric C. Kauffman, Heba W.Z. Khella, Ashley Orillion, W. Marston Linehan, and Khunsha Ahmed
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,Oncogene Proteins, Fusion ,Antineoplastic Agents ,Biology ,Article ,Deep sequencing ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Cell Line, Tumor ,Biomarkers, Tumor ,Animals ,Humans ,Gene silencing ,Gene Silencing ,Carcinoma, Renal Cell ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Regulation of gene expression ,Binding Sites ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,TOR Serine-Threonine Kinases ,Xenograft Model Antitumor Assays ,Kidney Neoplasms ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Insulin Receptor Substrate Proteins ,Cancer research ,TFEB ,Female ,Chromatin immunoprecipitation ,Protein Binding ,Signal Transduction - Abstract
Purpose: Translocation renal cell carcinoma (tRCC) represents a rare subtype of kidney cancer associated with various TFE3, TFEB, or MITF gene fusions that are not responsive to standard treatments for RCC. Therefore, the identification of new therapeutic targets represents an unmet need for this disease. Experimental Design: We have established and characterized a tRCC patient-derived xenograft, RP-R07, as a novel preclinical model for drug development by using next-generation sequencing and bioinformatics analysis. We then assessed the therapeutic potential of inhibiting the identified pathway using in vitro and in vivo models. Results: The presence of a SFPQ-TFE3 fusion [t(X;1) (p11.2; p34)] with chromosomal break-points was identified by RNA-seq and validated by RT-PCR. TFE3 chromatin immunoprecipitation followed by deep sequencing analysis indicated a strong enrichment for the PI3K/AKT/mTOR pathway. Consistently, miRNA microarray analysis also identified PI3K/AKT/mTOR as a highly enriched pathway in RP-R07. Upregulation of PI3/AKT/mTOR pathway in additional TFE3–tRCC models was confirmed by significantly higher expression of phospho-S6 (P < 0.0001) and phospho-4EBP1 (P < 0.0001) in established tRCC cell lines compared with clear cell RCC cells. Simultaneous vertical targeting of both PI3K/AKT and mTOR axis provided a greater antiproliferative effect both in vitro (P < 0.0001) and in vivo (P < 0.01) compared with single-node inhibition. Knockdown of TFE3 in RP-R07 resulted in decreased expression of IRS-1 and inhibited cell proliferation. Conclusions: These results identify TFE3/IRS-1/PI3K/AKT/mTOR as a potential dysregulated pathway in TFE3–tRCC, and suggest a therapeutic potential of vertical inhibition of this axis by using a dual PI3K/mTOR inhibitor for patients with TFE3–tRCC.
- Published
- 2018
- Full Text
- View/download PDF
17. Common ELF1 deletion in prostate cancer bolsters oncogenic ETS function, inhibits senescence and promotes docetaxel resistance
- Author
-
Peter C. Hollenhorst, Matthew J. Capone, Mary W. Ferris, and Justin A. Budka
- Subjects
0301 basic medicine ,Senescence ,chemotherapy resistance ,Cancer Research ,tumor suppressor ,Biology ,medicine.disease_cause ,law.invention ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,law ,Genetics ,medicine ,Gene ,Transcription factor ,Gene knockdown ,ELF1 ,Cell migration ,prostate cancer ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Suppressor ,Carcinogenesis ,Research Paper ,ETS - Abstract
ETS family transcription factors play major roles in prostate tumorigenesis with some acting as oncogenes and others as tumor suppressors. ETS factors can compete for binding at some cis-regulatory sequences, but display specific binding at others. Therefore, changes in expression of ETS family members during tumorigenesis can have complex, multimodal effects. Here we show that ELF1 was the most commonly down-regulated ETS factor in primary prostate tumors, and expression decreased further in metastatic disease. Genome-wide mapping in cell lines indicated that ELF1 has two distinct tumor suppressive roles mediated by distinct cis-regulatory sequences. First, ELF1 inhibited cell migration and epithelial-mesenchymal transition by interfering with oncogenic ETS functions at ETS/AP-1 cis-regulatory motifs. Second, ELF1 uniquely targeted and activated genes that promote senescence. Furthermore, knockdown of ELF1 increased docetaxel resistance, indicating that the genomic deletions found in metastatic prostate tumors may promote therapeutic resistance through loss of both RB1 and ELF1.
- Published
- 2018
- Full Text
- View/download PDF
18. EZH2 Modifies Sunitinib Resistance in Renal Cell Carcinoma by Kinome Reprogramming
- Author
-
Ashley Orillion, W. Andy Tao, Remi Adelaiye-Ogala, M Radovich, Giulio Draetta, Michael J. Buck, Peter C. Hollenhorst, Piergiorgio Pettazzoni, Janaiah Kota, Roberto Pili, Li Shen, Scott A. Persohn, Bradley A. Hancock, Justine V. Arrington, Brian P. McCarthy, Yong Zang, Kiersten Marie Miles, Joseph Irudayaraj, May Elbanna, Paul Territo, Mary W. Ferris, Mukund Seshadri, Eric Ciamporcero, Sreevani Arisa, Justin A. Budka, Heike Keilhack, Sreenivasulu Chintala, Chuan-Chih Hsu, and Nur P. Damayanti
- Subjects
Vascular Endothelial Growth Factor A ,0301 basic medicine ,Cancer Research ,Indoles ,Lung Neoplasms ,Antineoplastic Agents ,Mice, SCID ,Biology ,Article ,Mice ,03 medical and health sciences ,Cell Line, Tumor ,Sunitinib ,medicine ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,Pyrroles ,Kinome ,Phosphorylation ,Carcinoma, Renal Cell ,Mice, Inbred ICR ,Predictive marker ,Neovascularization, Pathologic ,EZH2 ,Receptor Protein-Tyrosine Kinases ,Cell cycle ,medicine.disease ,Xenograft Model Antitumor Assays ,Kidney Neoplasms ,Bevacizumab ,Clear cell renal cell carcinoma ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,Histone methyltransferase ,Cancer research ,Female ,Reprogramming ,medicine.drug - Abstract
Acquired and intrinsic resistance to receptor tyrosine kinase inhibitors (RTKi) represents a major hurdle in improving the management of clear cell renal cell carcinoma (ccRCC). Recent reports suggest that drug resistance is driven by tumor adaptation via epigenetic mechanisms that activate alternative survival pathways. The histone methyl transferase EZH2 is frequently altered in many cancers, including ccRCC. To evaluate its role in ccRCC resistance to RTKi, we established and characterized a spontaneously metastatic, patient-derived xenograft model that is intrinsically resistant to the RTKi sunitinib, but not to the VEGF therapeutic antibody bevacizumab. Sunitinib maintained its antiangiogenic and antimetastatic activity but lost its direct antitumor effects due to kinome reprogramming, which resulted in suppression of proapoptotic and cell-cycle–regulatory target genes. Modulating EZH2 expression or activity suppressed phosphorylation of certain RTKs, restoring the antitumor effects of sunitinib in models of acquired or intrinsically resistant ccRCC. Overall, our results highlight EZH2 as a rational target for therapeutic intervention in sunitinib-resistant ccRCC as well as a predictive marker for RTKi response in this disease. Cancer Res; 77(23); 6651–66. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
19. 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
20. 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
21. 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
22. Abstract 2366: HDAC inhibition improves immune checkpoint inhibitor efficacy in renal cell carcinoma
- Author
-
Justin A. Budka, Roberto Pili, and Nur P. Damayanti
- Subjects
Cancer Research ,Entinostat ,Tumor-infiltrating lymphocytes ,business.industry ,Cell cycle ,Natural killer cell ,chemistry.chemical_compound ,Immune system ,medicine.anatomical_structure ,Oncology ,chemistry ,medicine ,Myeloid-derived Suppressor Cell ,Cancer research ,business ,Vorinostat ,CD8 ,medicine.drug - Abstract
Background: Immune checkpoint inhibitors have shown clinical benefit in solid tumors, including renal cell carcinoma (RCC); however, the rate of clinical response remains modest and improved therapeutic approaches need to be tested. Growing evidence suggests that epigenetic modifying agents may have an immunomodulatory effect that improves the efficacy of immune checkpoint inhibitors. Our group has previously demonstrated that entinostat, a histone deacetylase (HDAC) inhibitor, decreases the function of regulatory T cells (Treg) and myeloid derived suppressor cells (MDSC), synergizing with PD-1 blockade. Here we assessed the combination of PD-1 blockade with pan-HDAC inhibition in a RCC model. Methods: To test the efficacy of combined PD-1 inhibition, mDX-400 (10 and 20 mg/kg I.P) (Merck & Co, Inc) with pan-HDAC inhibition, vorinostat (100 and 150 mg/kg I.P) (Merck & Co, Inc), we utilized a syngeneic mouse model of metastatic RCC following orthotopic implantation of RENCA cells in immunocompetent mice. Antitumor activity was assessed by measuring bioluminescence, end point tumor weights, and survival times. Immune profiling of tumor infiltrating lymphocytes (TILs) was performed by flow cytometry, immunohistochemistry, and immunofluorescence. Peripheral blood mononuclear cells (PBMC) were assessed for differential chromatin accessibility by Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq). Results: Significant reductions in tumor weight and lung metastases were observed in mice treated with the combination of vorinostat and mDX-400. Combination therapy significantly increased the survival of the mice (median survival = 60) compared to treatment with mDX-400 alone (median survival=42 days). Immune landscape profiling demonstrated an increase in natural killer cell infiltration (P=0.048) and decrease of exhausted T cells (P=0.049, CD8+ PD1+) in the combination group. Furthermore, decreased immunosuppressive Treg (CD4+ FOXP4+) and MDSC (CD11b+ Gr1+) populations were identified in the combination group. Analysis of the mouse PBMC ATAC-seq data in the combination and mDX400 alone conditions demonstrated numerous regions of differential chromatin accessibility. Pathway analysis of genes associated with increased accessibility in the combination treatment identified enrichment of cell cycle and immune activation pathways. Conclusions: Our results demonstrate that pan-HDAC inhibition augments the antitumor effect of immune checkpoint inhibitors, prolonging survival in our preclinical mouse model. This antitumor effect was achieved by changing the immune landscape in TILs and was associated with higher chromatin accessibility near genes involved in cell cycle progression and immune cell activation. Taken together, our results support the clinical testing of pan-HDAC inhibitors in combination with anti-PD-1. Citation Format: Justin Budka, Nur Damayanti, Roberto Pili. HDAC inhibition improves immune checkpoint inhibitor efficacy in renal cell carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2366.
- Published
- 2019
- Full Text
- View/download PDF
23. Abstract A184: T-cell rejuvenation is associated with vorinostat-induced immune response in combination with immune checkpoint blockade
- Author
-
Josue D. Ordaz, Xioana Chu, Justin A. Budka, Yue Wang, Ashley Orillion, Roberto Pili, Khunsha Ahmed, Nur P. Damayanti, and Yunlong Liu
- Subjects
Cancer Research ,Entinostat ,Tumor-infiltrating lymphocytes ,business.industry ,T cell ,medicine.medical_treatment ,Immunology ,Immune checkpoint ,Natural killer cell ,chemistry.chemical_compound ,medicine.anatomical_structure ,Immune system ,chemistry ,Cancer immunotherapy ,medicine ,Cancer research ,business ,Vorinostat ,medicine.drug - Abstract
Background: Immune checkpoint inhibitors targeting the PD-1/PD-L1 axis have shown clinical benefit in solid tumor patients, including renal cell carcinoma (RCC). However, the rate of clinical response remains modest. Growing evidence suggests that epigenetic modifying agents may have an immunomodulatory role. Our group has previously demonstrated that the selective class I histone deacetylase (HDAC) inhibitor entinostat decreases the function of regulatory T-cells (Treg) and myeloid-derived suppressor cells (MDSC), and synergizes with PD-1 blockade. In this study, we assessed the immunomodulatory activity and efficacy of combining PD-1 blockade with the pan-HDAC inhibitor vorinostat in a RCC model. Methods: To test the efficacy of a combination therapy with a PD-1 inhibitor, mDX-400 (10 and 20 mg/kg I.P) (Merck & Co., Inc.) and a pan-HDAC inhibitor, vorinostat (100 and 150 mg/kg I.P) (Merck & Co., Inc.), we utilized a syngeneic mouse model of metastatic RCC following orthotopic implantation of luciferase expressing RENCA cells in immunocompetent BALB/c mice. Antitumor activity was assessed by bioluminescence technique as well as end point measurements of tumor weights. Immune landscape profiling of tumor infiltrating lymphocytes (TILs) was performed by flow cytometry, immunohistochemistry, and immunofluorescence. Survival analysis was performed by Kaplan–Meier estimates and log-rank statistic. Differences in chromatin accessibility in peripheral blood mononuclear cells (PBMC) were assessed by Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq). Results: Statistically significant reductions in end point tumor weights, as well as lung metastases nodules, were observed in mice treated with the combination of vorinostat (100 mg/kg P=0.0391; 150 mg/kg P=0.0165) and mDX-400 (20 mg/kg) compared to vehicle, while no statistical significant reduction was observed in those treated with single-agent mDX-400. Combination therapy also significantly lengthened the survival of the mice (median survival = 60 days; P=0.009) compared to treatment with the single agent mDX-400 (median survival=42 days). Immune landscape profiling did not demonstrate a significant increase in CD8+ tumor infiltration (P=0.479), but a statistically significant increase in natural killer cell infiltration (P=0.048) was observed. Though the CD8+ tumor infiltration was unchanged, a significant reduction (P=0.049) of exhausted CD8+ T-cells (CD8+PD1+) was observed in the combination treatment compared to mDX400 alone. Furthermore, a decrease was observed in the immunosuppressive Tregs (CD4+FOXP4+) and MDSC (CD11b+Gr1+) in the combination group compared to mDX400 alone. Bioinformatic analyses of ATAC-seq data from the PBMC cells of mice in the combination treatment and mDX400 alone showed increased chromatin accessibility between the two conditions. Pathway analysis of genes associated with more accessible chromatin in the combination treatment than mDX400 treatment identified enrichment of cell cycle control and immune cell activation pathways. Conclusions: Our results demonstrate that the pan-HDAC inhibitor vorinostat augments the antitumor effect of immune checkpoint inhibitor mDX-400 and prolongs survival in the RENCA model. This combination advantage was achieved by changing the immune landscape in TILs, especially by decreasing the exhausted subset of T-cells. The combination of these drugs is associated with higher chromatin accessibility near genes involved in cell cycle progression and immune cell activation. Taken together, our results support the clinical testing of pan-HDAC inhibitors in combination of PD-1 inhibitors and provide a novel potential immunomodulatory effect of epigenetic drugs. Citation Format: Nur P. Damayanti, Justin A. Budka, Josue D. Ordaz, Ashley Orillion, Khunsha Ahmed, Xioana Chu, Yue Wang, Yunlong Liu, Roberto Pili. T-cell rejuvenation is associated with vorinostat-induced immune response in combination with immune checkpoint blockade [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A184.
- Published
- 2019
- Full Text
- View/download PDF
24. Abstract A023: Regulation of ERG function in prostate cells by phosphorylation and interaction with Ewing’s sarcoma breakpoint protein EWS
- Author
-
Peter C. Hollenhorst, Vivekananda Kedage, Brady G. Strittmatter, Taylor R. Nicholas, Travis J. Jerde, Nagarathinam Selvaraj, and Justin A. Budka
- Subjects
Cancer Research ,genetic structures ,biology ,EZH2 ,macromolecular substances ,medicine.disease ,TMPRSS2 ,eye diseases ,ETV1 ,Prostate cancer ,Oncology ,Mitogen-activated protein kinase ,Coactivator ,Cancer research ,medicine ,biology.protein ,Phosphorylation ,sense organs ,Transcription factor - Abstract
More than one-half of prostate tumors have a chromosomal rearrangement that results in the overexpression of an oncogenic ETS family transcription factor. The most common fusion, TMPRSS2/ERG, results in expression of ERG, a protein that is not normally expressed in prostate epithelia. When ERG is expressed in prostate cells it is thought to bind to enhancer elements and regulate gene expression by recruiting transcriptional coactivators and/or corepressors. We have recently shown that the EWS protein acts as a coactivator for ERG in prostate cells and this interaction is required for ERG-mediated xenograft tumor growth. Interestingly, the EWS interaction may be the key requirement that separates oncogenic from nononcogenic ETS factors, as EWS only interacts with four ETS family members involved in prostate cancer gene rearrangements (ERG, ETV1, ETV4, and ETV5), but not with other ETS family members. This ETS/EWS interaction also indicates a common molecular mechanism involved in prostate cancer and Ewing’s sarcoma, a cancer caused by gene fusions that express chimeric EWS/ETS proteins. ERG also interacts with corepressors such as EZH2, a subunit of PRC2. We have recently found that the interaction between ERG and EZH2/PRC2 is regulated by a series of phosphorylation events on ERG. The MAP kinase ERK can bind a high-affinity docking sequence in ERG, resulting in phosphorylation of a nearby serine, S215. This phosphorylation event leads to a conformational change in ERG that allows ERK to phosphorylate a second serine, S96. S96 phosphorylation then disrupts the interaction between ERG and EZH2/PRC2, allowing ERG to activate gene expression. Together, the interaction of ERG with EWS and the regulation of ERG function by ERK-mediated phosphorylation, represent molecular mechanisms that could serve as targets for therapeutic intervention in ERG-positive prostate cancer. Citation Format: Vivekananda Kedage, Taylor R. Nicholas, Brady G. Strittmatter, Nagarathinam Selvaraj, Justin A. Budka, Travis J. Jerde, Peter C. Hollenhorst. Regulation of ERG function in prostate cells by phosphorylation and interaction with Ewing’s sarcoma breakpoint protein EWS [abstract]. In: Proceedings of the AACR Special Conference: Prostate Cancer: Advances in Basic, Translational, and Clinical Research; 2017 Dec 2-5; Orlando, Florida. Philadelphia (PA): AACR; Cancer Res 2018;78(16 Suppl):Abstract nr A023.
- Published
- 2018
- Full Text
- View/download PDF
25. Prostate cancer ETS rearrangements switch a cell migration gene expression program from RAS/ERK to PI3K/AKT regulation
- Author
-
Travis J. Jerde, Nagarathinam Selvaraj, Justin A. Budka, Mary W. Ferris, and Peter C. Hollenhorst
- Subjects
MAPK/ERK pathway ,Male ,Cancer Research ,Blotting, Western ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Transduction, Genetic ,Anti-apoptotic Ras signalling cascade ,Cell Line, Tumor ,PTEN ,Humans ,Cell migration ,HRAS ,Extracellular Signal-Regulated MAP Kinases ,Protein kinase B ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Gene Rearrangement ,0303 health sciences ,PI3K/AKT ,Prostate cancer ,biology ,Proto-Oncogene Proteins c-ets ,Reverse Transcriptase Polymerase Chain Reaction ,Research ,Prostatic Neoplasms ,Gene rearrangement ,RAS/ERK ,Elafin ,Gene Expression Regulation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,ras Proteins ,Molecular Medicine ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Signal Transduction ,ETS - Abstract
Background The RAS/ERK and PI3K/AKT pathways induce oncogenic gene expression programs and are commonly activated together in cancer cells. Often, RAS/ERK signaling is activated by mutation of the RAS or RAF oncogenes, and PI3K/AKT is activated by loss of the tumor suppressor PTEN. In prostate cancer, PTEN deletions are common, but, unlike other carcinomas, RAS and RAF mutations are rare. We have previously shown that over-expression of “oncogenic” ETS transcription factors, which occurs in about one-half of prostate tumors due to chromosome rearrangement, can bypass the need for RAS/ERK signaling in the activation of a cell migration gene expression program. In this study we test the role of RAS/ERK and PI3K/AKT signaling in the function of oncogenic ETS proteins. Results We find that oncogenic ETS expression negatively correlates with RAS and RAF mutations in prostate tumors. Furthermore, the oncogenic ETS transcription factors only increased cell migration in the absence of RAS/ERK activation. In contrast to RAS/ERK, it has been reported that oncogenic ETS expression positively correlates with PI3K/AKT activation. We identified a mechanistic explanation for this finding by showing that oncogenic ETS proteins required AKT signaling to activate a cell migration gene expression program through ETS/AP-1 binding sequences. Levels of pAKT correlated with the ability of oncogenic ETS proteins to increase cell migration, but this process did not require mTORC1. Conclusions Our findings indicate that oncogenic ETS rearrangements cause a cell migration gene expression program to switch from RAS/ERK control to PI3K/AKT control and provide a possible explanation for the high frequency of PTEN, but not RAS/RAF mutations in prostate cancer.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.