33 results on '"Vijaya L. Dommeti"'
Search Results
2. Supplemental Figure Legends from BET Bromodomain Inhibitors Enhance Efficacy and Disrupt Resistance to AR Antagonists in the Treatment of Prostate Cancer
- Author
-
Arul M. Chinnaiyan, Felix Y. Feng, Shaomeng Wang, Nora M. Navone, Stephen R. Plymate, June Escara-Wilke, Ingrid J. Apel, Pranathi M. Krishnamurthy, Vijaya L. Dommeti, Kari Wilder-Romans, and Irfan A. Asangani
- Abstract
Supplemental Figure Legends
- Published
- 2023
- Full Text
- View/download PDF
3. Supplemental Figure 2 from BET Bromodomain Inhibitors Enhance Efficacy and Disrupt Resistance to AR Antagonists in the Treatment of Prostate Cancer
- Author
-
Arul M. Chinnaiyan, Felix Y. Feng, Shaomeng Wang, Nora M. Navone, Stephen R. Plymate, June Escara-Wilke, Ingrid J. Apel, Pranathi M. Krishnamurthy, Vijaya L. Dommeti, Kari Wilder-Romans, and Irfan A. Asangani
- Abstract
Direct comparison of various BET inhibitors in prostate cancer cells.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplemental Figure 3 from BET Bromodomain Inhibitors Enhance Efficacy and Disrupt Resistance to AR Antagonists in the Treatment of Prostate Cancer
- Author
-
Arul M. Chinnaiyan, Felix Y. Feng, Shaomeng Wang, Nora M. Navone, Stephen R. Plymate, June Escara-Wilke, Ingrid J. Apel, Pranathi M. Krishnamurthy, Vijaya L. Dommeti, Kari Wilder-Romans, and Irfan A. Asangani
- Abstract
De-recruitment of BRD2/3/4 from the SRSF1 and U2AF1 promoter by JQ1.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplemental Information from BET Bromodomain Inhibitors Enhance Efficacy and Disrupt Resistance to AR Antagonists in the Treatment of Prostate Cancer
- Author
-
Arul M. Chinnaiyan, Felix Y. Feng, Shaomeng Wang, Nora M. Navone, Stephen R. Plymate, June Escara-Wilke, Ingrid J. Apel, Pranathi M. Krishnamurthy, Vijaya L. Dommeti, Kari Wilder-Romans, and Irfan A. Asangani
- Abstract
Supplemental Materials and Methods
- Published
- 2023
- Full Text
- View/download PDF
6. Supplemental Tables 1 and 2 from BET Bromodomain Inhibitors Enhance Efficacy and Disrupt Resistance to AR Antagonists in the Treatment of Prostate Cancer
- Author
-
Arul M. Chinnaiyan, Felix Y. Feng, Shaomeng Wang, Nora M. Navone, Stephen R. Plymate, June Escara-Wilke, Ingrid J. Apel, Pranathi M. Krishnamurthy, Vijaya L. Dommeti, Kari Wilder-Romans, and Irfan A. Asangani
- Abstract
Antibodies and PCR Primers
- Published
- 2023
- Full Text
- View/download PDF
7. Supplemental Figure 4 from BET Bromodomain Inhibitors Enhance Efficacy and Disrupt Resistance to AR Antagonists in the Treatment of Prostate Cancer
- Author
-
Arul M. Chinnaiyan, Felix Y. Feng, Shaomeng Wang, Nora M. Navone, Stephen R. Plymate, June Escara-Wilke, Ingrid J. Apel, Pranathi M. Krishnamurthy, Vijaya L. Dommeti, Kari Wilder-Romans, and Irfan A. Asangani
- Abstract
AR- and ERG-positive PDX model displays sensitivity to JQ1.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplemental Figure 1 from BET Bromodomain Inhibitors Enhance Efficacy and Disrupt Resistance to AR Antagonists in the Treatment of Prostate Cancer
- Author
-
Arul M. Chinnaiyan, Felix Y. Feng, Shaomeng Wang, Nora M. Navone, Stephen R. Plymate, June Escara-Wilke, Ingrid J. Apel, Pranathi M. Krishnamurthy, Vijaya L. Dommeti, Kari Wilder-Romans, and Irfan A. Asangani
- Abstract
Enzalutamide resistant VCaP prostate cancer cells.
- Published
- 2023
- Full Text
- View/download PDF
9. MP01-05 REVEALING NEW PARADIGMS IN MALE INFERTILITY THROUGH THE LENS OF STRESS-INDUCED GRANULES
- Author
-
Joel E. Berends, Vijaya L. Dommeti, Jeffrey N. Dudley, Stephanie Carras, Catherine S. Nam, Tyler Robinson, Evan Keller, James M. Dupree, and Sethu Pitchiaya
- Subjects
Urology - Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Data from EWS/ETS-Driven Ewing Sarcoma Requires BET Bromodomain Proteins
- Author
-
Irfan A. Asangani, Felix Y. Feng, Zhaohui S. Qin, Tianlei Xu, Ingrid J. Apel, Archana Nallasivam, Pranathi M. Krishnamurthy, Vijaya L. Dommeti, Carl G. Engelke, Ramakrishnan Natesan, Kari Wilder-Romans, Aishwarya Pawar, and Paradesi Naidu Gollavilli
- Abstract
Supplementary Data from EWS/ETS-Driven Ewing Sarcoma Requires BET Bromodomain Proteins
- Published
- 2023
- Full Text
- View/download PDF
11. Data from EWS/ETS-Driven Ewing Sarcoma Requires BET Bromodomain Proteins
- Author
-
Irfan A. Asangani, Felix Y. Feng, Zhaohui S. Qin, Tianlei Xu, Ingrid J. Apel, Archana Nallasivam, Pranathi M. Krishnamurthy, Vijaya L. Dommeti, Carl G. Engelke, Ramakrishnan Natesan, Kari Wilder-Romans, Aishwarya Pawar, and Paradesi Naidu Gollavilli
- Abstract
The EWS/ETS fusion transcription factors drive Ewing sarcoma (EWS) by orchestrating an oncogenic transcription program. Therapeutic targeting of EWS/ETS has been unsuccessful; however, identifying mediators of the EWS/ETS function could offer new therapeutic options. Here, we describe the dependency of EWS/ETS-driven transcription upon chromatin reader BET bromdomain proteins and investigate the potential of BET inhibitors in treating EWS. EWS/FLI1 and EWS/ERG were found in a transcriptional complex with BRD4, and knockdown of BRD2/3/4 significantly impaired the oncogenic phenotype of EWS cells. RNA-seq analysis following BRD4 knockdown or inhibition with JQ1 revealed an attenuated EWS/ETS transcriptional signature. In contrast to previous reports, JQ1 reduced proliferation and induced apoptosis through MYC-independent mechanisms without affecting EWS/ETS protein levels; this was confirmed by depleting BET proteins using PROTAC-BET degrader (BETd). Polycomb repressive complex 2 (PRC2)-associated factor PHF19 was downregulated by JQ1/BETd or BRD4 knockdown in multiple EWS lines. EWS/FLI1 bound a distal regulatory element of PHF19, and EWS/FLI1 knockdown resulted in downregulation of PHF19 expression. Deletion of PHF19 via CRISPR-Cas9 resulted in a decreased tumorigenic phenotype, a transcriptional signature that overlapped with JQ1 treatment, and increased sensitivity to JQ1. PHF19 expression was also associated with worse prognosis in patients with EWS. In vivo, JQ1 demonstrated antitumor efficacy in multiple mouse xenograft models of EWS. Together these results indicate that EWS/ETS requires BET epigenetic reader proteins for its transcriptional program and can be mitigated by BET inhibitors. This study provides a clear rationale for the clinical utility of BET inhibitors in treating EWS.Significance:These findings reveal the dependency of EWS/ETS transcription factors on BET epigenetic reader proteins and demonstrate the potential of BET inhibitors for the treatment of EWS. Cancer Res; 78(16); 4760–73. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
12. Argonaute 2 modulates EGFR-RAS signaling to promote mutant
- Author
-
Ronald F, Siebenaler, Seema, Chugh, Jessica J, Waninger, Vijaya L, Dommeti, Carson, Kenum, Malay, Mody, Anudeeta, Gautam, Nidhi, Patel, Alec, Chu, Pushpinder, Bawa, Jennifer, Hon, Richard D, Smith, Heather, Carlson, Xuhong, Cao, John J G, Tesmer, Sunita, Shankar, and Arul M, Chinnaiyan
- Abstract
Activating mutations in RAS GTPases drive nearly 30% of all human cancers. Our prior work described an essential role for Argonaute 2 (AGO2), of the RNA-induced silencing complex, in mutant
- Published
- 2022
13. AGO2 promotes tumor progression in KRAS-driven mouse models of non-small cell lung cancer
- Author
-
Rui Wang, Fengyun Su, Alice Xu, Arul M. Chinnaiyan, Yunhui Cheng, Sunita Shankar, Jennifer Hon, Lisha Wang, Rahul Mannan, Jean Ching-Yi Tien, Andrew Goodrum, Vijaya L. Dommeti, Seema Chugh, Xuhong Cao, Xiaoming Wang, Carson Kenum, and Yuping Zhang
- Subjects
Lung Neoplasms ,Medical Sciences ,AGO2 ,Proliferative index ,MAP Kinase Signaling System ,Biology ,Malignancy ,medicine.disease_cause ,Proto-Oncogene Proteins p21(ras) ,Mice ,Carcinoma, Non-Small-Cell Lung ,medicine ,KRAS ,Gene silencing ,Animals ,Allele ,Lung cancer ,neoplasms ,Multidisciplinary ,Argonaute ,Biological Sciences ,medicine.disease ,digestive system diseases ,respiratory tract diseases ,Mice, Inbred C57BL ,Disease Models, Animal ,Tumor progression ,Argonaute Proteins ,Cancer research ,Disease Progression ,Signal Transduction ,nonsmall cell lung cancer - Abstract
Significance RAS proteins (HRAS, NRAS, and KRAS) integrate extracellular trophic signals to promote cell proliferation. Constitutively active KRAS drives tumor initiation and progression in nonsmall cell lung cancer (NSCLC). As RAS proteins are often refractory to direct pharmacological inhibition, RAS-interacting proteins are under investigation as potential drug targets. We previously found Argonaute 2 (AGO2) to bind RAS and positively regulate RAS-dependent signaling pathways. AGO2 knockdown blunted proliferation in NSCLC cells containing a KRAS-activating mutation. Here, we demonstrate that AGO2 promotes tumor progression in multiple mouse models of KRAS-driven NSCLC. In these animals, Ago2 knockout impairs tumor growth, lowers pathologic grade, and inhibits KRAS signaling. Targeting the AGO2-KRAS interaction may hold future therapeutic promise in NSCLC and other KRAS-driven malignancies., Lung cancer is the deadliest malignancy in the United States. Non–small cell lung cancer (NSCLC) accounts for 85% of cases and is frequently driven by activating mutations in the gene encoding the KRAS GTPase (e.g., KRASG12D). Our previous work demonstrated that Argonaute 2 (AGO2)—a component of the RNA-induced silencing complex (RISC)—physically interacts with RAS and promotes its downstream signaling. We therefore hypothesized that AGO2 could promote KRASG12D-dependent NSCLC in vivo. To test the hypothesis, we evaluated the impact of Ago2 knockout in the KPC (LSL-Kras G12D/+;p53f/f;Cre) mouse model of NSCLC. In KPC mice, intratracheal delivery of adenoviral Cre drives lung-specific expression of a stop-floxed KRASG12D allele and biallelic ablation of p53. Simultaneous biallelic ablation of floxed Ago2 inhibited KPC lung nodule growth while reducing proliferative index and improving pathological grade. We next applied the KPHetC model, in which the Clara cell–specific CCSP-driven Cre activates KRASG12D and ablates a single p53 allele. In these mice, Ago2 ablation also reduced tumor size and grade. In both models, Ago2 knockout inhibited ERK phosphorylation (pERK) in tumor cells, indicating impaired KRAS signaling. RNA sequencing (RNA-seq) of KPC nodules and nodule-derived organoids demonstrated impaired canonical KRAS signaling with Ago2 ablation. Strikingly, accumulation of pERK in KPC organoids depended on physical interaction of AGO2 and KRAS. Taken together, our data demonstrate a pathogenic role for AGO2 in KRAS-dependent NSCLC. Given the prevalence of this malignancy and current difficulties in therapeutically targeting KRAS signaling, our work may have future translational relevance.
- Published
- 2021
14. Abstract 2538: Role of Argonaute 2 in regulation of immune microenvironment in pancreatic cancer
- Author
-
Jennifer Hon, Carson Kenum, Pushpinder S. Bawa, Vijaya L. Dommeti, Anastasia A. Sahu, Miriam Gandham, Chi-Chiang Li, Zainab I. Taher, Sylvia Zelenka-Wang, Jean C. Tien, Sunita Shankar, Seema Chugh, and Arul M. Chinnaiyan
- Subjects
Cancer Research ,Oncology - Abstract
KRAS mutations have been observed in nearly 95% of pancreatic ductal adenocarcinoma (PDAC), however targeting KRAS remains a major therapeutic challenge. Previous studies from our group have discovered a novel interaction between KRAS and Argonaute 2 (AGO2) and have uncovered a significant role of this interaction in regulating KRAS signaling. Knockout of AGO2 in genetically engineered mouse models of KRAS driven pancreatic and lung cancer dramatically impacted tumor progression. Intriguingly, in pancreatic cancer loss of AGO2 expression resulted in early pancreatic intraepithelial lesions (PanINs) that failed to progress to PDAC. We observed increased senescence in these AGO2 knockout lesions that abrogated PDAC progression. Further, there was pronounced infiltration of immune cells in pancreata lacking AGO2 in comparison to wild type. We particularly observed a 20-fold increase in natural killer (NK) cells population in pancreata lacking AGO2. This instigated us to evaluate potential role of AGO2 in regulating immune microenvironment in pancreatic cancer. Gene set enrichment analysis of AGO2 knockout SW1990 PDAC cells revealed significant upregulation of inflammatory and interferon response pathways. We observed increased expression of several signaling proteins implicated in immune activation pathways. Further characterization of these knockout cells indicated upregulation of MHC proteins that consequently added to mechanistic insights. Additionally, we are also exploring syngeneic models of pancreatic cancer and we will present findings from our ongoing studies to evaluate the impact of AGO2 knockout on immune axis. Overall, our findings suggest potential involvement of AGO2 in regulating immune microenvironment in pancreatic cancer. Citation Format: Jennifer Hon, Carson Kenum, Pushpinder S. Bawa, Vijaya L. Dommeti, Anastasia A. Sahu, Miriam Gandham, Chi-Chiang Li, Zainab I. Taher, Sylvia Zelenka-Wang, Jean C. Tien, Sunita Shankar, Seema Chugh, Arul M. Chinnaiyan. Role of Argonaute 2 in regulation of immune microenvironment in pancreatic cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2538.
- Published
- 2022
- Full Text
- View/download PDF
15. An essential role for Argonaute 2 in EGFR-KRAS signaling in pancreatic cancer development
- Author
-
Ronald F. Siebenaler, Jean Ching-Yi Tien, Vijaya L. Dommeti, Rahul Mannan, Stephanie J. Ellison, Sethuramasundaram Pitchiaya, Xuhong Cao, Seema Chugh, Howard C. Crawford, Ingrid J. Apel, Jessica Waninger, Chandan Kumar-Sinha, Andrew Goodrum, Sanjana Eyunni, Sylvia Zelenka-Wang, Pankaj Vats, Yuping Zhang, Malay Mody, Jiaqi Shi, Xiaoming Wang, Alice Xu, Sunita Shankar, John J.G. Tesmer, and Arul M. Chinnaiyan
- Subjects
0301 basic medicine ,Male ,endocrine system diseases ,Mutant ,General Physics and Astronomy ,medicine.disease_cause ,Metastasis ,Mice ,0302 clinical medicine ,Phosphorylation ,lcsh:Science ,Cellular Senescence ,Multidisciplinary ,Argonaute ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Argonaute Proteins ,Disease Progression ,Female ,KRAS ,Protein Binding ,Signal Transduction ,Senescence ,Genotype ,Science ,Mice, Transgenic ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Pancreatic cancer ,Cell Line, Tumor ,microRNA ,medicine ,Animals ,Humans ,Cancer models ,Alleles ,Cell Membrane ,General Chemistry ,medicine.disease ,digestive system diseases ,Pancreatic Neoplasms ,030104 developmental biology ,Cancer research ,lcsh:Q ,Tumor Suppressor Protein p53 ,Neoplasm Transplantation - Abstract
Both KRAS and EGFR are essential mediators of pancreatic cancer development and interact with Argonaute 2 (AGO2) to perturb its function. Here, in a mouse model of mutant KRAS-driven pancreatic cancer, loss of AGO2 allows precursor lesion (PanIN) formation yet prevents progression to pancreatic ductal adenocarcinoma (PDAC). Precursor lesions with AGO2 ablation undergo oncogene-induced senescence with altered microRNA expression and EGFR/RAS signaling, bypassed by loss of p53. In mouse and human pancreatic tissues, PDAC progression is associated with increased plasma membrane localization of RAS/AGO2. Furthermore, phosphorylation of AGO2Y393 disrupts both the wild-type and oncogenic KRAS-AGO2 interaction, albeit under different conditions. ARS-1620 (G12C-specific inhibitor) disrupts the KRASG12C-AGO2 interaction, suggesting that the interaction is targetable. Altogether, our study supports a biphasic model of pancreatic cancer development: an AGO2-independent early phase of PanIN formation reliant on EGFR-RAS signaling, and an AGO2-dependent phase wherein the mutant KRAS-AGO2 interaction is critical for PDAC progression., Argonaute 2 (AGO2) binds RAS and is required for cellular transformation. Here, the authors establish a KRAS-driven mouse model of pancreatic cancer with conditional loss of AGO2 and show that the early phase of neoplastic lesion initiation is dependent on EGFR/RAS but not AGO2, while AGO2 is required for pancreatic ductal adenocarcinoma progression and metastasis.
- Published
- 2020
16. EWS/ETS-Driven Ewing Sarcoma Requires BET Bromodomain Proteins
- Author
-
Carl G. Engelke, Tianlei Xu, Zhaohui S. Qin, Pranathi Meda Krishnamurthy, Archana Nallasivam, Aishwarya Pawar, Felix Y. Feng, Ramakrishnan Natesan, Irfan A. Asangani, Kari Wilder-Romans, Vijaya L. Dommeti, Ingrid J. Apel, and Paradesi Naidu Gollavilli
- Subjects
0301 basic medicine ,Cancer Research ,BRD4 ,Oncogene Proteins, Fusion ,Transcription, Genetic ,Apoptosis ,Cell Cycle Proteins ,Sarcoma, Ewing ,Epigenesis, Genetic ,Mice ,03 medical and health sciences ,Cell Line, Tumor ,Animals ,Humans ,Epigenetics ,Transcription factor ,Cell Proliferation ,Regulation of gene expression ,Gene knockdown ,biology ,Proto-Oncogene Protein c-fli-1 ,Polycomb Repressive Complex 2 ,Nuclear Proteins ,Xenograft Model Antitumor Assays ,Bromodomain ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,FLI1 ,biology.protein ,Cancer research ,RNA-Binding Protein EWS ,PRC2 ,Transcription Factors - Abstract
The EWS/ETS fusion transcription factors drive Ewing sarcoma (EWS) by orchestrating an oncogenic transcription program. Therapeutic targeting of EWS/ETS has been unsuccessful; however, identifying mediators of the EWS/ETS function could offer new therapeutic options. Here, we describe the dependency of EWS/ETS-driven transcription upon chromatin reader BET bromdomain proteins and investigate the potential of BET inhibitors in treating EWS. EWS/FLI1 and EWS/ERG were found in a transcriptional complex with BRD4, and knockdown of BRD2/3/4 significantly impaired the oncogenic phenotype of EWS cells. RNA-seq analysis following BRD4 knockdown or inhibition with JQ1 revealed an attenuated EWS/ETS transcriptional signature. In contrast to previous reports, JQ1 reduced proliferation and induced apoptosis through MYC-independent mechanisms without affecting EWS/ETS protein levels; this was confirmed by depleting BET proteins using PROTAC-BET degrader (BETd). Polycomb repressive complex 2 (PRC2)-associated factor PHF19 was downregulated by JQ1/BETd or BRD4 knockdown in multiple EWS lines. EWS/FLI1 bound a distal regulatory element of PHF19, and EWS/FLI1 knockdown resulted in downregulation of PHF19 expression. Deletion of PHF19 via CRISPR-Cas9 resulted in a decreased tumorigenic phenotype, a transcriptional signature that overlapped with JQ1 treatment, and increased sensitivity to JQ1. PHF19 expression was also associated with worse prognosis in patients with EWS. In vivo, JQ1 demonstrated antitumor efficacy in multiple mouse xenograft models of EWS. Together these results indicate that EWS/ETS requires BET epigenetic reader proteins for its transcriptional program and can be mitigated by BET inhibitors. This study provides a clear rationale for the clinical utility of BET inhibitors in treating EWS. Significance: These findings reveal the dependency of EWS/ETS transcription factors on BET epigenetic reader proteins and demonstrate the potential of BET inhibitors for the treatment of EWS. Cancer Res; 78(16); 4760–73. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
17. Abstract 2936: Ago2 induces Kras signaling and promotes tumor progression in mouse models of lung adenocarcinoma
- Author
-
Fengyu Su, Alice Xu, Sunita Shankar, Yuping Zhang, Xiaoming Wang, Arul M. Chinnaiyan, Rahul Mannan, Jean C. Tien, Vijaya L. Dommeti, Lisha Wang, Xuhong Cao, Yunhui Cheng, Andrew Goodrum, and Seema Chugh
- Subjects
Cancer Research ,Lung ,business.industry ,medicine.disease_cause ,medicine.disease ,respiratory tract diseases ,medicine.anatomical_structure ,Oncology ,Tumor progression ,Cancer research ,Medicine ,Adenocarcinoma ,KRAS ,business - Abstract
Lung cancer is the deadliest malignancy in the United States. Non-small cell lung cancer (NSCLC) accounts for 85% of cases, and is frequently driven by activating mutations in the gene encoding the KRAS GTPase (e.g. KRASG12D). Our previous work demonstrated that Argonaute 2 (AGO2)—a component of the RNA induced silencing complex (RISC)—physically interacts with KRAS and stabilizes it at the protein level. In multiple cell lines, AGO2 knockdown reduces KRAS protein level and attenuates cell proliferation. We therefore hypothesized that AGO2 could promote KRASG12D-dependent NSCLC in vivo. To test the hypothesis, we evaluated the impact of Ago2 knockout in the KPC mouse model of NSCLC. In KPC mice, intratracheal delivery of adenoviral Cre drives lung-specific expression of a stop-floxed KRASG12D allele and biallelic ablation of p53. Simultaneous biallelic ablation of floxed Ago2 inhibited KPC lung nodule growth while reducing proliferative index and improving pathological grade. We next applied the KPHetC model, in which the clara cell-specific CCSP-driven Cre activates KRASG12D and ablates a single p53 allele. In these mice, Ago2 ablation also reduced tumor size and grade. In both models, Ago2 knockout inhibited ERK phosphorylation in tumor nodules, indicating impaired KRAS signaling. Furthermore, RNA-seq analysis of KPC nodules demonstrated broadly reduced expression of Kras-related genes. Taken together, our data demonstrate a novel pathogenic role for AGO2 in KRAS-dependent NSCLC. Given the prevalence of this malignancy and current difficulties in therapeutically targeting KRAS signaling, our work has future translational relevance. Citation Format: Jean C. Tien, Seema Chugh, Andrew E. Goodrum, Yunhui Cheng, Lisha Wang, Rahul Mannan, Xiaoming Wang, Vijaya L. Dommeti, Yuping Zhang, Alice Xu, Fengyu Su, Xuhong Cao, Sunita Shankar, Arul M. Chinnaiyan. Ago2 induces Kras signaling and promotes tumor progression in mouse models of lung adenocarcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 2936.
- Published
- 2021
- Full Text
- View/download PDF
18. BET Bromodomain Inhibitors Enhance Efficacy and Disrupt Resistance to AR Antagonists in the Treatment of Prostate Cancer
- Author
-
Stephen R. Plymate, Vijaya L. Dommeti, Ingrid J. Apel, Irfan A. Asangani, Arul M. Chinnaiyan, Kari Wilder-Romans, Shaomeng Wang, Pranathi Meda Krishnamurthy, Felix Y. Feng, June Escara-Wilke, and Nora M. Navone
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Cell Survival ,medicine.drug_class ,Biology ,Antiandrogen ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,Prostate ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,Nitriles ,Phenylthiohydantoin ,Androgen Receptor Antagonists ,medicine ,Animals ,Humans ,Enzalutamide ,Molecular Biology ,Cell Proliferation ,Cancer ,Azepines ,Triazoles ,medicine.disease ,Xenograft Model Antitumor Assays ,Bromodomain ,Androgen receptor ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,chemistry ,Drug Resistance, Neoplasm ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Benzamides ,Cancer research ,Acetanilides ,Heterocyclic Compounds, 3-Ring ,Signal Transduction - Abstract
Next-generation antiandrogen therapies, such as enzalutamide and abiraterone, have had a profound impact on the management of metastatic castration-resistant prostate cancer (mCRPC). However, mCRPC patients invariably develop resistance to these agents. Here, a series of clonal cell lines were developed from enzalutamide-resistant prostate tumor xenografts to study the molecular mechanism of resistance and test their oncogenic potential under various treatment conditions. Androgen receptor (AR) signaling was maintained in these cell lines, which acquired potential resistance mechanisms, including expression of AR-variant 7 (AR-v7) and glucocorticoid receptor. BET bromodomain inhibitors were shown previously to attenuate AR signaling in mCRPC; here, we demonstrate the efficacy of bromodomain and extraterminal (BET) inhibitors in enzalutamide-resistant prostate cancer models. AR antagonists, enzalutamide, and ARN509 exhibit enhanced prostate tumor growth inhibition when combined with BET inhibitors, JQ1 and OTX015, respectively. Taken together, these data provide a compelling preclinical rationale to combine BET inhibitors with AR antagonists to subvert resistance mechanisms. Implications: Therapeutic combinations of BET inhibitors and AR antagonists may enhance the clinical efficacy in the treatment of mCRPC. Visual Overview: http://mcr.aacrjournals.org/content/14/4/324/F1.large.jpg. Mol Cancer Res; 14(4); 324–31. ©2016 AACR.
- Published
- 2016
- Full Text
- View/download PDF
19. Abstract 2577: AGO2 interaction limits wild type RAS activation yet essential for disease progression in oncogenic KRAS driven cancers
- Author
-
Vijaya L. Dommeti, Howard C. Crawford, Jiaqi Shi, Andrew Goodrum, Sylvia Zelenka-Wang, Ingrid J. Apel, Sanjana Eyunni, Ronald F. Siebenaler, Sethuramsundaram Pitchiaya, Sunita Shankar, Yuping Zhang, Xiaoming Wang, Javed Siddiqui, Jean Ching-Yi Tien, Arul M. Chinnaiyan, Kristin M. Juckette, Seema Chugh, Jessica Waninger, Chandan Kumar-Sinha, and Xuhong Cao
- Subjects
MAPK/ERK pathway ,Cancer Research ,Wild type ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,Oncology ,Pancreatic cancer ,Cancer research ,medicine ,SOS1 ,KRAS ,Carcinogenesis ,PI3K/AKT/mTOR pathway - Abstract
KRAS mutations drive over 30% of all cancers and 90% of pancreatic cancer. To investigate additional potential modulators of RAS-mediated oncogenesis, we previously performed a screen for direct interactors of RAS in a panel of cancer cell lines and identified a direct interaction between KRAS and Argonaute 2 (AGO2), independent of KRAS mutation status. To define the effects of conditional loss of AGO2 in KRASG12D-driven cancer, we employed a genetically engineered mouse model of pancreatic cancer (KC model). Genetic ablation of AGO2 did not interfere with development of the normal pancreas or KRASG12D-driven early precursor pancreatic intraepithelial neoplasia (PanIN) lesions. However, AGO2 loss prevented progression from early to late PanIN lesions, development of pancreatic ductal adenocarcinoma (PDAC), and metastatic progression. This resulted in increased of KRASG12D mutant mice deficient in AGO2 expression. Mechanistically, lack of PanIN to PDAC progression was due to oncogene-induced senescence (OIS) through activation of EGFR-wild type RASWT-phospho ERK signaling including reduced baseline autophagy in mice with AGO2 loss. Interestingly, in the KPC model simultaneous expression of oncogenic KRAS and loss of a p53 allele abrogates this phenotype, suggesting that p53 deficiency overcomes the OIS check by AGO2. However, in both mouse and human PDAC, AGO2 expression increased with disease progression and, interestingly, was predominant at the plasma membrane, where it co-localized with RAS. These in vivo analyses support a biphasic model of pancreatic cancer development: an AGO2-independent early phase of PanIN formation reliant on EGFR and WT RAS signaling, and an AGO2-dependent phase wherein the mutant KRAS-AGO2 interaction is critical for PDAC progression. In the KRASG12D driven lung cancer mouse model, we see a similar requirement for AGO2 for diseases progression–but not for initiation–despite p53 loss indicating a context dependent requirement for AGO2. Given that AGO2 binds both the mutant and the WT forms of KRAS, we also investigated the role of RASWT-AGO2 interaction. Analysis using purified proteins showed that AGO2 competes with SOS1 to limit RASWT activation. AGO2 had no effect on the intrinsic GTPase activity of RAS or NF1-mediated GAP activity. In line with this data, AGO2 null mouse embryonic fibroblasts showed increased RAS activity and activation of MAPK/PI3K pathways. Further, using cell line models, we also showed that phosphorylation of AGO2Y393 by EGFR disrupted the interaction of RASWT with AGO2 at the membrane, but did not affect the interaction of mutant KRAS with AGO2. On the other hand, ARS-1620, a G12C-specific inhibitor, disrupted the KRASG12C-AGO2 interaction specifically in cells harboring this mutant, demonstrating that the oncogenic KRAS-AGO2 interaction can be pharmacologically targeted. Altogether, our findings reveal that the AGO2 interaction regulates RASWT and is essential for mutant KRAS driven oncogenesis. Citation Format: Sunita Shankar, Jean Ching-Yi Tien, Ronald F. Siebenaler, Seema Chugh, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Jessica Waninger, Kristin M. Juckette, Xiao-Ming Wang, Sanjana Eyunni, Andrew Goodrum, Yuping Zhang, Ingrid J. Apel, Javed Siddiqui, Xuhong Cao, Jiaqi Shi, Sethuramsundaram Pitchiaya, Chandan Kumar-Sinha, Howard C. Crawford, Arul M. Chinnaiyan. AGO2 interaction limits wild type RAS activation yet essential for disease progression in oncogenic KRAS driven cancers [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 2577.
- Published
- 2020
- Full Text
- View/download PDF
20. Abstract B39: Characterization of the interaction between KRAS and Argonaute 2
- Author
-
Ronald F. Siebenaler, Jean Tien, Heather A. Carlson, Vijaya L. Dommeti, Richard D. Smith, Arul M. Chinnaiyan, Jessica Waninger, Malay Mody, Chandan Kumar-Sinha, John J.G. Tesmer, Tyler S. Beyett, and Sunita Shankar
- Subjects
Cancer Research ,Oncology ,Chemistry ,medicine ,KRAS ,Computational biology ,Argonaute ,medicine.disease_cause ,Molecular Biology - Abstract
KRAS is one of three members of the RAS family of small GTPases, and nearly one third of all human cancers harbor mutations in the KRAS gene. These mutations lead to a constitutively active GTP-bound form of KRAS that results in aberrant activation of MAPK and PI3K pathways that control cell differentiation, growth, and survival. While much has been learned about the effectors of KRAS and its complex signaling network, little progress has been made in successfully targeting it in the treatment of cancer. Recently, we identified a functional protein-protein interaction between KRAS and Argonaute 2 (AGO2), a core component of the RNA-induced silencing complex (RISC). Investigation of this interaction revealed that the switch II domain of KRAS (both wild-type [WT] and mutant) binds to the N-terminal (Nt) domain of AGO2. Mutant KRAS-AGO2 binding results in attenuation of the RNAi function of AGO2 by preventing microRNA unwinding, a necessary step in the formation of the mature RISC complex (model 1). Analysis of this interaction revealed that KRAS-AGO2 binding is direct through Tyr64 of KRAS and Lys112/Glu114 of AGO2. Recently, using genetically engineered mouse models (GEMM), we showed that mutant KRAS is dependent on AGO2 for the development of pancreatic ductal adenocarcinoma (PDAC) from early precursor lesions in the pancreas, suggesting a critical role for the KRAS-AGO2 interaction in vivo. To further characterize KRAS-AGO2 binding, we purified recombinant proteins individually, isolated complex via size exclusion chromatography (SEC), performed differential scanning fluorimetry (DSF), isothermal titration calorimetry (ITC), and small angle x-ray scattering (SAXS). Additionally, in order to evaluate endogenous regulators of the KRAS-AGO2 interaction we investigated the role of the epidermal growth factor receptor (EGFR), which is known to regulate the RNAi function of AGO2 via phosphorylation at Tyr393. Interestingly, we observed that phosphorylation at this residue disrupts the interaction of AGO2 with WT but not mutant KRAS, providing a basis for differential regulation of the KRAS-AGO2 interaction through growth factor activation. Finally, to examine structural changes induced by EGFR phosphorylation of AGO2 at Tyr393 that may affect KRAS binding, we performed parallel molecular dynamics simulations (MDS) of nonphosphorylated and phospho-Tyr393 AGO2. These studies revealed immediate structural reorganization of the RAS binding site and an outward rotation of the entire Nt domain of AGO2. Taken together, these data shed light on a potential mechanism underlying the differential regulation of mutant vs. WT KRAS. Citation Format: Jessica Waninger, Sunita Shankar, Ronald Siebenaler, Malay Mody, Jean Tien, Vijaya Dommeti, Tyler Beyett, Richard Smith, Chandan Kumar-Sinha, Heather Carlson, John Tesmer, Arul Chinnaiyan. Characterization of the interaction between KRAS and Argonaute 2 [abstract]. In: Proceedings of the AACR Special Conference on Targeting RAS-Driven Cancers; 2018 Dec 9-12; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Res 2020;18(5_Suppl):Abstract nr B39.
- Published
- 2020
- Full Text
- View/download PDF
21. Abstract A21: Loss of Argonaute 2 leads to oncogene-induced senescence in mutant RAS-driven cancer
- Author
-
Ronald F. Siebenaler, Sylvia Zelenka-Wang, Malay Mody, Vijaya L. Dommeti, Sunita Shankar, Arul M. Chinnaiyan, Jean C. Tien, Jessica Waninger, Chandan Kumar-Sinha, and Seema Chugh
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Cancer Research ,Mutant ,Cancer ,Argonaute ,Biology ,medicine.disease ,medicine.disease_cause ,Oncology ,Pancreatic cancer ,medicine ,Cancer research ,Gene silencing ,HRAS ,KRAS ,Molecular Biology - Abstract
The RAS gene family is among the most commonly mutated genes within cancer, but little progress has been made in successfully targeting RAS mutations. Targeting binding partners of mutated RAS, however, presents a promising alternative therapeutic strategy. With the goal of uncovering novel interactors of RAS, we recently identified Argonaute 2 (AGO2) of the RNA-induced silencing complex (RISC) as a novel partner of the Switch II domain of KRAS. In order to assess the role of AGO2 in KRAS-G12D driven disease, we developed a mouse model of pancreatic cancer with conditional loss of AGO2. While AGO2 knockout did not prevent development of early precursor pancreatic intraepithelial (PanIN) lesions, loss of AGO2 prevented progression to late-stage PanINs, pancreactic ductal adenocarcinoma (PDAC), and metastatic disease. AGO2 null lesions displayed increased activation of the EGFR-RAS signaling axis during PanIN development. This signaling resulted in an increase in WT RAS-GTP activation, pEGFR-Y1068, and pERK levels leading to the development of oncogene-induced senescence in these PanIN lesions. Furthermore, we observed that EGFR-mediated phosphorylation of AGO2-Y393 disrupted the interaction between WT RAS and AGO2. This regulation by EGFR, however, was blocked in cells expressing mutant KRAS. These results suggested that the interaction of mutant RAS and AGO2 was vital to tumor development. To better assess the role of AGO2 loss in mutant RAS driven cancer, we performed AGO2 knockdown in multiple cell lines expressing mutations in either NRAS or HRAS isoforms. In each cell line, AGO2 directly interacted with KRAS, NRAS, and KRAS. In addition to suppressing growth in mutant RAS-driven cells (T24: HRAS-G12V, SK-MEL-2: NRAS-Q61H), loss of AGO2 produced marked increases in beta-galactosidase and p16 expression, as well as a decrease in cyclin D1, suggesting development of oncogene-induced senescence. Interestingly, upon AGO2 loss, cells displayed induction of pEGFR and pERK similar to what was observed in our pancreatic mouse model, and despite decreased expression of mutant RAS, WT RAS-GTP loading upon AGO2 loss was strongly induced. Together these results suggest a unique EGFR-AGO2-RAS signaling axis that requires AGO2-RAS interaction to prevent induction of oncogene-induced senescence in mutant RAS-driven cancers. Citation Format: Ronald F. Siebenaler, Sunita Shankar, Jean C. Tien, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Jessica Waninger, Malay Mody, Seema Chugh, Chandan Kumar-Sinha, Arul M. Chinnaiyan. Loss of Argonaute 2 leads to oncogene-induced senescence in mutant RAS-driven cancer [abstract]. In: Proceedings of the AACR Special Conference on Targeting RAS-Driven Cancers; 2018 Dec 9-12; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Res 2020;18(5_Suppl):Abstract nr A21.
- Published
- 2020
- Full Text
- View/download PDF
22. Abstract A20: An essential role for Argonaute 2 in mouse models of KRAS driven cancers
- Author
-
Jiaqi Shi, Andrew Goodrum, Vijaya L. Dommeti, John J.G. Tesmer, Alice Xu, Kristin M. Juckette, Sunita Shankar, Arul M. Chinnaiyan, Sanjana Eyunni, Xiaoming Wang, Malay Mody, Ronald F. Siebenaler, Yuping Zhang, Javed Siddiqui, Sylvia Zelenka-Wang, Grace Tsaloff, Jean Ching-Yi Tien, Xuhong Cao, Ingrid J. Apel, Richard D. Smith, Jessica Waninger, Chandan Kumar-Sinha, Lisha Wang, Seema Chugh, and Heather A. Carlson
- Subjects
Cancer Research ,Pancreatic Intraepithelial Neoplasia ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,Oncology ,Growth factor receptor ,Pancreatic cancer ,medicine ,Cancer research ,Adenocarcinoma ,Gene silencing ,KRAS ,Carcinogenesis ,Molecular Biology - Abstract
In 2016, we identified a direct interaction between RAS and Argonaute 2 (AGO2), a key mediator of RNA-mediated gene silencing that is required for KRAS-driven oncogenesis using pancreatic and lung cancer cell line models. Recently, we employed the genetically engineered mouse model of pancreatic cancer to define the effects of conditional loss of AGO2 in KRASG12D driven pancreatic cancer. Genetic ablation of AGO2 did not interfere with development of the normal pancreas or KRASG12D-driven early precursor pancreatic intraepithelial neoplasia (PanIN) lesions. However, AGO2 loss prevents progression from early to late PanIN lesions, development of pancreatic ductal adenocarcinoma (PDAC), and metastatic progression. This results in a dramatic increase in survival of KRASG12D mutant mice deficient in AGO2 expression. Using validated pan-RAS and AGO2 antibodies for immunofluorescence (IF) and proximity ligation assay (PLA), we observed increased RAS and AGO2 co-localization at the plasma membrane in mouse and human pancreatic tissues associated with PDAC progression. AGO2 ablation permits PanIN initiation driven by the EGFR-RAS axis; however rather than progressing to PDAC, these lesions undergo profound oncogene-induced senescence (OIS). Since PanIN development requires EGFR and is not AGO2 dependent, we probed the effects of EGF stimulation in cell lines expressing wild-type and mutant forms of KRAS (using co-IP and PLA analyses). In wild-type RAS expressing cells, grown in media containing serum, RAS-AGO2 co-localization was limited to the intracellular regions of the cells, which dramatically increased and shifted to the plasma membrane under conditions of stress (serum starvation). Interestingly, EGF stimulation disrupted this membrane RAS-AGO2 interaction and restored it to intracellular basal levels. Using phosphorylation-deficient AGO2 mutants, we demonstrate that the disruption of wild type-RAS-AGO2 interaction is due to AGO2Y393 phosphorylation, a target of EGFR. Interestingly, the mutant KRAS-AGO2 interaction is not subject to EGFR activation, suggesting that although both the wild-type and mutant RAS bind AGO2, they are differentially regulated through growth factor receptor activation. Taken together, our study supports a biphasic model of pancreatic cancer development: an AGO2-independent early phase of PanIN formation reliant on EGFR and wild-type RAS signaling, and an AGO2-dependent phase wherein the mutant KRAS-AGO2 interaction is critical for PDAC progression. In the lung cancer mouse model, we also observed a similar dependence of AGO2 in KRAS-driven lung adenocarcinoma. Along with related abstracts detailing the mechanisms of OIS mediated by AGO2 (Ronald Siebenaler) and evidence of direct interaction between oncogenic KRAS and AGO2 with an affinity of 200nM (Jessica Waninger), we present our latest studies related to the KRAS-AGO2 interaction. Citation Format: Sunita Shankar, Jean Ching-Yi Tien, Ronald F. Siebenaler, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Jessica Waninger, Xiao-Ming Wang, Kristin M. Juckette, Alice Xu, Seema Chugh, Malay Mody, Sanjana Eyunni, Andrew Goodrum, Grace Tsaloff, Yuping Zhang, Ingrid J. Apel, Lisha Wang, Javed Siddiqui, Richard D. Smith, Heather A. Carlson, John Tesmer, Xuhong Cao, Jiaqi Shi, Chandan Kumar-Sinha, Arul M. Chinnaiyan. An essential role for Argonaute 2 in mouse models of KRAS driven cancers [abstract]. In: Proceedings of the AACR Special Conference on Targeting RAS-Driven Cancers; 2018 Dec 9-12; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Res 2020;18(5_Suppl):Abstract nr A20.
- Published
- 2020
- Full Text
- View/download PDF
23. An Essential Role for Argonaute 2 in EGFR-KRAS Signaling in Pancreatic Cancer Development
- Author
-
Javed Siddiqui, Xiaoming Wang, Jiaqi Shi, Heather A. Carlson, Lisha Wang, Vijaya L. Dommeti, Ingrid J. Apel, Alice Xu, Seema Chugh, Grace Tsaloff, Sunita Shankar, Richard D. Smith, Sylvia Zelenka-Wang, John J.G. Tesmer, Jessica Waninger, Chandan Kumar-Sinha, Arul M. Chinnaiyan, Ronald F. Siebenaler, Andrew Goodrum, Howard C. Crawford, Sanjana Eyunni, Malay Mody, Kristin M. Juckette, Yuping Zhang, Jean Ching-Yi Tien, and Xuhong Cao
- Subjects
endocrine system diseases ,Mutant ,Pancreatic Intraepithelial Neoplasia ,Regulator ,Biology ,Argonaute ,medicine.disease ,medicine.disease_cause ,digestive system diseases ,Metastasis ,Pancreatic cancer ,Cancer research ,medicine ,Gene silencing ,KRAS - Abstract
KRAS and EGFR are known essential mediators of pancreatic cancer development. In addition, KRAS and EGFR have both been shown to interact with and perturb the function of Argonaute 2 (AGO2), a key regulator of RNA-mediated gene silencing. Here, we employed a genetically engineered mouse model of pancreatic cancer to define the effects of conditional loss ofAGO2inKRASG12D-driven pancreatic cancer. Genetic ablation ofAGO2does not interfere with development of the normal pancreas orKRASG12D-driven early precursor pancreatic intraepithelial neoplasia (PanIN) lesions. Remarkably, however,AGO2is required for progression from early to late PanIN lesions, development of pancreatic ductal adenocarcinoma (PDAC), and metastasis.AGO2ablation permits PanIN initiation driven by the EGFR-RAS axis, but rather than progressing to PDAC, these lesions undergo profound oncogene-induced senescence (OIS). Loss ofTrp53(p53) in this model obviates the requirement ofAGO2for PDAC development. In mouse and human pancreatic tissues, increased expression of AGO2 and elevated co-localization with RAS at the plasma membrane is associated with PDAC progression. Furthermore, phosphorylation of AGO2Y393by EGFR disrupts the interaction of wild-type RAS with AGO2 at the membrane, but does not affect the interaction of mutant KRAS with AGO2. ARS-1620, a G12C-specific inhibitor, disrupts the KRASG12C-AGO2 interaction specifically in pancreatic cancer cells harboring this mutant, demonstrating that the oncogenic KRAS-AGO2 interaction can be pharmacologically targeted. Taken together, our study supports a biphasic model of pancreatic cancer development: anAGO2-independent early phase of PanIN formation reliant on EGFR-RAS signaling, and anAGO2-dependent phase wherein the mutant KRAS-AGO2 interaction is critical to prevent OIS in PanINs and allow progression to PDAC.
- Published
- 2017
- Full Text
- View/download PDF
24. Abstract 94: Role of Argonaute 2 in oncogene induced senescence in a pancreatic cancer mouse model
- Author
-
Ronald F. Siebenaler, Sanjana Eyunni, Vijaya L. Dommeti, Kristin M. Juckette, Arul M. Chinnaiyan, Sylvia Z. Wang, Sunita Shankar, Lisha Wang, Seema Chugh, and Jean C. Tien
- Subjects
Senescence ,Cancer Research ,Gene knockdown ,Wild type ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,medicine.anatomical_structure ,Oncology ,Pancreatic cancer ,Cancer research ,medicine ,KRAS ,Pancreas ,Survival rate - Abstract
Pancreatic cancer is a highly devastating malignancy with a very poor survival rate of 7%. Mutations in KRAS have been identified in more than 90% of PDAC patients. Previous work from our lab has shown that KRAS directly interacts with Argonaute 2 (AGO2) to promote cellular transformation. To study the involvement of AGO2 in KRASG12D-driven cancers, AGO2 expression was ablated in the KrasG12D/+; p48 Cre model (KC) model of pancreatic cancer. AGO2 floxed mice (AGO2loxP/loxP) were crossed with KC mice, resulting in oncogenic KRAS expression along with knockout of AGO2 in pancreatic acinar cells. Survival and disease progression were compared between wild-type (AGO2+/+; KRASG12D; p48Cre), heterozygous (AGO2fl/+; KRASG12D; p48Cre), and homozygous (AGO2fl/fl; KRASG12D; p48Cre) experimental mice groups. Homozygous knockout of AGO2 in KC mice resulted in significantly increased survival as compared to wild type and heterozygous mice. Pancreatic ductal adenocarcinoma (PDAC) and metastases were restricted to the wild-type and heterozygous mice. Pancreas from AGO2fl/fl; KRASG12D; p48Cre mice develop only early pancreatic intraepithelial lesions (PanINs), which fail to progress to PDAC. Senescence-associated β-galactosidase staining showed strong and significant increase in senescence in PanIN lesions mice lacking AGO2 expression as compared to AGO2+/+; KRASG12D; p48Cre mice. This suggests that AGO2 prevents oncogene induced senescence (OIS) as a result of KRASG12D expression and allows PanIN to PDAC progression. To gain mechanistic insights of OIS due to AGO2 loss, we evaluated markers for OIS including p16, p53, p21, gamma γH2AX, and RAS-associated signaling (pERK and pAkt). Analysis of PanIN lesions lacking AGO2 showed increased p16 levels and high levels of phospho-ERK, compared to PDAC from pancreas with AGO2 expression. In order to extend these observations in cell line models, we performed AGO2 knockdown in T24 cells harboring HRASG12V. Surprisingly, cells with low AGO2 levels underwent OIS, which was similar to the pancreatic mouse model and was accompanied with increased phospho-ERK signaling and p16 expression. Further studies are underway to determine the contribution of the RAS-AGO2 interaction in the development of OIS. Additionally, we are using CRISPR/Cas9 technology to screen pancreatic cancer cell lines with AGO2 knockout for their dependence on AGO2 and their ability to undergo OIS in the absence of AGO2 expression. We will present findings from our ongoing studies involving the role of AGO2 loss in the KPC (KRASG12D; p53fl/+; Cre) model, wherein OIS will be assessed in the absence of p53, a canonical inducer of cellular senescence. Citation Format: Seema Chugh, Jean C. Tien, Ronald F. Siebenaler, Vijaya L. Dommeti, Sylvia Z. Wang, Sanjana Eyunni, Kristin M. Juckette, Lisha Wang, Sunita Shankar, Arul M. Chinnaiyan. Role of Argonaute 2 in oncogene induced senescence in a pancreatic cancer mouse model [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 94.
- Published
- 2019
- Full Text
- View/download PDF
25. Abstract 957: An essential role for Argonaute 2 in mouse models of KRAS-driven cancers
- Author
-
Heather A. Carlson, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Seema Chugh, Sanjana Eyunni, Malay Mody, Kristin M. Juckette, Ingrid J. Apel, Grace Tsaloff, Jiaqi Shi, Alice Xu, Arul M. Chinnaiyan, Richard D. Smith, John J.G. Tesmer, Jessica Waninger, Chandan Kumar-Sinha, Ronald F. Siebenaler, Xiaoming Wang, Andrew Goodrum, Javed Siddiqui, Jean Ching-Yi Tien, Xuhong Cao, Sunita Shankar, and Yuping Zhang
- Subjects
Cancer Research ,Pancreatic Intraepithelial Neoplasia ,Wild type ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,Oncology ,Growth factor receptor ,Pancreatic cancer ,Cancer research ,medicine ,Gene silencing ,KRAS ,Carcinogenesis - Abstract
In 2016, we identified a direct interaction between RAS and Argonaute 2 (AGO2), a key mediator of RNA-mediated gene silencing, that is essential for KRAS-driven oncogenesis using pancreatic and lung cancer cell line models. Recently, we employed a genetically engineered mouse model of pancreatic cancer to define the effects of conditional loss of AGO2 in KRASG12D-driven pancreatic cancer (KC model). Genetic ablation of AGO2 did not interfere with development of the normal pancreas or KRASG12D-driven early precursor pancreatic intraepithelial neoplasia (PanIN) lesions. However, AGO2 loss prevents progression from early to late PanIN lesions, development of pancreatic ductal adenocarcinoma (PDAC), and metastatic progression. This results in a dramatic increase in the survival of KRASG12D mutant mice deficient in AGO2 expression. Mechanistically, lack of PanIN to PDAC progression was due to oncogene-induced senescence (OIS) through activation of EGFR-wild type RAS-phosphoERK signaling in the absence of AGO2. Using validated pan-RAS and AGO2 antibodies for immunofluorescence (IF) and proximity ligation assay (PLA), we observed increased RAS and AGO2 co-localization at the plasma membrane in mouse and human pancreatic tissues associated with PDAC progression. While AGO2 ablation permits PanIN initiation driven by the EGFR-RAS axis, these lesions undergo OIS rather than progressing to PDAC. Further, we used co-IP and PLA analyses to probe the effects of EGF stimulation in cell lines expressing wild-type and mutant forms of KRAS. In wild-type RAS expressing cells, RAS-AGO2 co-localization and interaction were limited to the intracellular regions of the cells, and dramatically increased and shifted to the plasma membrane under conditions of stress (serum starvation). Interestingly, EGF stimulation disrupted this membrane RAS-AGO2 interaction and restored it to intracellular levels. Using phosphorylation-deficient AGO2 mutants, we further demonstrate that the disruption of wild-type RAS-AGO2 interaction is due to EGFR-mediated AGO2Y393 phosphorylation. Interestingly, mutant KRAS-AGO2 interaction is not subject to EGFR activation, suggesting that although both the wild type and mutant RAS bind AGO2, they are differentially regulated through growth factor receptor activation. We will discuss our ongoing studies evaluating the effects of AGO2 ablation in the KRASG12Ddriven lung cancer mouse model and PDAC progression with p53 loss (KPC model). Our recent in vivo work supports a biphasic model of pancreatic cancer development: an AGO2-independent early phase of PanIN formation reliant on EGFR and wild-type RAS signaling, and an AGO2-dependent phase wherein the mutant KRAS-AGO2 interaction is critical for PDAC progression. Citation Format: Sunita Shankar, Jean Ching-Yi Tien, Ronald F. Siebenaler, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Jessica Waninger, Kristin M. Juckette, Alice Xu, Xiao-Ming Wang, Seema Chugh, Malay Mody, Sanjana Eyunni, Andrew Goodrum, Grace Tsaloff, Yuping Zhang, Ingrid J. Apel, Javed Siddiqui, Richard D. Smith, Heather A. Carlson, John Tesmer, Xuhong Cao, Jiaqi Shi, Chandan Kumar-Sinha, Arul M. Chinnaiyan. An essential role for Argonaute 2 in mouse models of KRAS-driven cancers [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 957.
- Published
- 2019
- Full Text
- View/download PDF
26. Abstract 956: An essential role for Argonaute 2 in EGFR-KRAS signaling in pancreatic cancer development
- Author
-
Ronald F. Siebenaler, Sunita Shankar, Jean C. Tien, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Seema Chugh, Ingrid J. Apel, Malay Mody, Anudeeta Gautam, Chandan Kumar-Sinha, and Arul M. Chinnaiyan
- Subjects
Cancer Research ,Oncology - Abstract
The RAS gene family is among the most commonly mutated genes within cancer, but little progress has been made in successfully targeting RAS mutations. Targeting binding partners of mutated RAS presents as a promising alternative therapeutic strategy. With the goal of uncovering novel interactors of RAS, we recently identified Argonaute 2 (AGO2) of the RNA-induced silencing complex (RISC) as a novel partner of the KRAS through its Switch II domain. In order to assess the role of AGO2 in KRASG12D driven disease, we developed a mouse model of pancreatic cancer with conditional loss of AGO2. While AGO2 knockout did not prevent development of early precursor pancreatic intraepithelial (PanIN) lesions, AGO2 null lesions displayed increased activation of the EGFR-RAS signaling axis during PanIN development that failed to progress to late stage PanINs, pancreatic ductal adenocarcinoma (PDAC), and metastatic disease. This resulted in a dramatic increase in the survival of mice with AGO2 ablation. Unlike the PanINs in AGO2 sufficient mice, the early PanIN lesions with AGO2 ablation showed staining for the senescence associated beta galactosidase activity, suggesting that AGO2 loss induces oncogene induced senescence. To extend these observations and explore the role of AGO2 interaction with mutant forms of HRAS and NRAS proteins, we performed co-IP of AGO2 with RAS proteins using isoform specific antibodies. Both HRAS and NRAS bound AGO2 in T24 cells (HRASG12V) and SK-MEL-2 cells (NRASQ61H), respectively. In T24 cells, AGO2 knockdown led to the senescent phenotype and was accompanied with changes in the EGFR-RAS signaling axis, similar to that observed in the PanINs of the mice with AGO2 loss. In this cell line model, AGO2 loss reduced mutant HRAS expression and increased wild type RAS activity. These signaling effects were also consistent with our observation that AGO2 loss increased RAS activation in the mouse embryonic fibroblast (MEF) model. Together with our previous work with mutant KRAS dependent cells, these data suggest that 1) AGO2-wild type RAS binding prevents RAS activation and 2) mutant RAS-AGO2 association regulates oncogenic RAS levels in cell line models. Studies on the mouse model and the close proximity of RAS and AGO2 with EGFR also furthered our understanding of the RAS-AGO2 interaction. Using a variety of cell line models, we observed that EGFR-mediated phosphorylation of AGO2Y393 disrupts the interaction between WT RAS and AGO2. However, the mutant KRAS-AGO2 interaction was recalcitrant to EGFR regulation. This provides the first instance of a nucleotide dependent association of RAS and AGO2 and sheds light on the dynamic nature of the RAS-AGO2 interaction. Citation Format: Ronald F. Siebenaler, Sunita Shankar, Jean C. Tien, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Seema Chugh, Ingrid J. Apel, Malay Mody, Anudeeta Gautam, Chandan Kumar-Sinha, Arul M. Chinnaiyan. An essential role for Argonaute 2 in EGFR-KRAS signaling in pancreatic cancer development [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 956.
- Published
- 2019
- Full Text
- View/download PDF
27. Abstract 3020: An essential role for Argonaute 2 in EGFR-KRAS signaling in pancreatic cancer development
- Author
-
Sunita Shankar, Jean Tien, Ronald F. Siebenaler, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Kristin M. Juckette, Alice Xu, Malay Mody, Andrew Goodrum, Grace Tsaloff, Ingrid J. Apel, Lisha Wang, Javed Siddiqui, Jiaqi Shi, Chandan Kumar-Sinha, and Arul Chinnaiyan
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Cancer Research ,Wild type ,Cancer ,Biology ,Argonaute ,medicine.disease ,medicine.disease_cause ,Oncology ,Pancreatic cancer ,Cancer research ,medicine ,Gene silencing ,KRAS ,HRAS - Abstract
The RAS gene family is among the most commonly mutated genes within cancer, but little progress has been made in successfully targeting RAS mutations. Targeting binding partners of mutated RAS presents as a promising alternative therapeutic strategy. With the goal of uncovering novel interactors of RAS, we recently identified Argonaute 2 (AGO2) of the RNA-induced silencing complex (RISC) as a novel partner of the KRAS through its Switch II domain. In order to assess the role of AGO2 in KRASG12D driven disease, we developed a mouse model of pancreatic cancer with conditional loss of AGO2. While AGO2 knockout did not prevent development of early precursor pancreatic intraepithelial (PanIN) lesions, AGO2 null lesions displayed increased activation of the EGFR-RAS signaling axis during PanIN development that failed to progress to late stage PanINs, pancreatic ductal adenocarcinoma (PDAC), and metastatic disease. This resulted in a dramatic increase in the survival of mice with AGO2 ablation. Unlike the PanINs in AGO2 sufficient mice, the early PanIN lesions with AGO2 ablation showed staining for the senescence associated beta galactosidase activity, suggesting that AGO2 loss induces oncogene induced senescence. To extend these observations and explore the role of AGO2 interaction with mutant forms of HRAS and NRAS proteins, we performed co-IP of AGO2 with RAS proteins using isoform specific antibodies. Both HRAS and NRAS bound AGO2 in T24 cells (HRASG12V) and SK-MEL-2 cells (NRASQ61H), respectively. In T24 cells, AGO2 knockdown led to the senescent phenotype and was accompanied with changes in the EGFR-RAS signaling axis, similar to that observed in the PanINs of the mice with AGO2 loss. In this cell line model, AGO2 loss reduced mutant HRAS expression and increased wild type RAS activity. These signaling effects were also consistent with our observation that AGO2 loss increased RAS activation in the mouse embryonic fibroblast (MEF) model. Together with our previous work with mutant KRAS dependent cells, these data suggest that 1) AGO2-wild type RAS binding prevents RAS activation and 2) mutant RAS-AGO2 association regulates oncogenic RAS levels in cell line models. Studies on the mouse model and the close proximity of RAS and AGO2 with EGFR also furthered our understanding of the RAS-AGO2 interaction. Using a variety of cell line models, we observed that EGFR-mediated phosphorylation of AGO2Y393 disrupts the interaction between WT RAS and AGO2. However, the mutant KRAS-AGO2 interaction was recalcitrant to EGFR regulation. This provides the first instance of a nucleotide dependent association of RAS and AGO2 and sheds light on the dynamic nature of the RAS-AGO2 interaction. Citation Format: Ronald F. Siebenaler, Sunita Shankar, Jean C. Tien, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Seema Chugh, Ingrid J. Apel, Malay Mody, Anudeeta Gautam, Chandan Kumar-Sinha, Arul M. Chinnaiyan. An essential role for Argonaute 2 in EGFR-KRAS signaling in pancreatic cancer development [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 956.
- Published
- 2018
- Full Text
- View/download PDF
28. Abstract 4370: Regulation of AGO2-KRAS interaction through epidermal growth factor receptor
- Author
-
Ronald F. Siebenaler, Jean C. Tien, Arul M. Chinnaiyan, Sunita Shankar, Chandan Kumar-Sinha, Malay Mody, and Vijaya L. Dommeti
- Subjects
MAPK/ERK pathway ,Cancer Research ,biology ,medicine.disease_cause ,Oncology ,Cancer research ,biology.protein ,medicine ,Gene silencing ,Phosphorylation ,Epidermal growth factor receptor ,KRAS ,Carcinogenesis ,Protein kinase B ,PI3K/AKT/mTOR pathway - Abstract
The RAS gene family is among the most commonly mutated genes within cancer. While much research has elucidated its major functions and downstream pathways, little progress has been made in successfully targeting RAS mutations. We recently identified Argonaute 2 (AGO2) of the RNA-induced silencing complex (RISC) as a novel partner of the Switch II domain of KRAS. We have found that stable knockdown of AGO2 in KRAS-dependent cell lines lead to a decrease in KRAS protein expression with a subsequent decrease in cellular proliferation. In addition, we observed a decrease in microRNA unwinding in the presence of mutant KRAS, suggesting this interaction inhibits the endogenous RNAi function of AGO2. Despite this clear connection between KRAS and AGO2 in KRAS mediated oncogenesis, the precise function of this interaction remains unclear in both normal and cancer biology. In order to identify endogenous regulators of AGO2-RAS, we investigated the ability of EGFR signaling to modulate the AGO2-RAS interaction. We established two mouse embryonic fibroblast cell lines (NIH-3T3 and MEF) with complete knockout of AGO2. When compared to normal control cells, we found that knockout of AGO2 resulted in an increase in WT RAS-GTP activation levels, phosphorylation of Y1068-EGFR, and MAPK/ERK and PI3K/AKT signaling. Rescue of AGO2 knockout resulted in a return to normal levels of active RAS-GTP, pEGFR, and downstream signaling. Recent studies have described EGFR phosphorylation of AGO2 under hypoxic cell conditions, resulting in the inhibition of AGO2 association with RISC members. In order to better characterize the relationship between EGFR-AGO2-RAS, we found that overnight serum starvation followed by stimulation with EGF led to a decrease in AGO2-RAS co-IP in WT KRAS cells. Blocking AGO2 phosphorylation with a Y393F mutant of AGO2 prevented AGO2-RAS dissociation following EGFR stimulation. While WT KRAS cell lines displayed regulation of AGO2-RAS via EGFR, the phosphorylation of AGO2Y393 was unable to disrupt the interaction of AGO2 with mutant KRAS following stimulation with EGF. Together these results suggest a unique EGFR-AGO2-RAS signaling axis, and its dysregulation by mutant KRAS could increase oncogenic growth through promotion of AGO2-RAS interaction in cancer. Citation Format: Ronald F. Siebenaler, Sunita Shankar, Jean C. Tien, Vijaya L. Dommeti, Malay Mody, Chandan Kumar-Sinha, Arul M. Chinnaiyan. Regulation of AGO2-KRAS interaction through epidermal growth factor receptor [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4370.
- Published
- 2018
- Full Text
- View/download PDF
29. Abstract 3029: An in vivo role for Argonaute 2 in KRAS driven pancreatic cancer
- Author
-
Arul M. Chinnaiyan, Jean Tien, Sunita Shankar, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Chandan Kumar-Sinha, Jiaqi Shi, and Ingrid A. Apel
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Argonaute ,medicine.disease ,medicine.disease_cause ,In vivo ,Internal medicine ,Pancreatic cancer ,medicine ,Cancer research ,KRAS ,business - Abstract
Oncogenic mutations in RAS provide a compelling yet intractable therapeutic target. Using co-immunoprecipitation mass spectrometry, we recently identified an interaction between RAS and Argonaute 2 (AGO2), the core component of the RNA silencing machinery exerting translational control of mRNA transcripts. RAS and AGO2 co-sediment and co-localize in intracellular endomembrane bound organelles. The AGO2 N-terminal domain directly binds the KRAS Switch II region, irrespective of GDP/GTP bound to RAS. Functionally, we observed that AGO2 was required for maximal oncogenic KRAS levels and AGO2 knock-down attenuates cell proliferation in mutant KRAS-dependent cells. Intriguingly, AGO2 mediated microRNA processing (RISC activity) was attenuated in cancer cells expressing mutant KRAS compared to those expressing wild type RAS. Yet, these initial investigations using cell line models do not reveal a clear role for AGO2 in KRAS driven oncogenesis. Therefore, we probed the role of AGO2 in the well-established pancreatic ductal adenocarcinoma (PDAC) mouse model driven by oncogenic KRAS. Towards this end, we generated LSL-KrasG12D; Pft1a Cre; AGO2flox/flox (KCA) mice. Preliminary analysis (8-10 weeks of age) suggests that compared to LSL-KRASG12D (KC) mice, homozygous loss of AGO2 results in increased KRAS driven Acinar to Ductal Metaplasia (ADM) phenotype considered as precursors of PanIN (pancreatic intraepithelial neoplasia) and PDAC. This mirrors the phenotype of loss of Dicer in the same model further reinforcing a role for microRNAs in restraining KRAS oncogenic programs. Yet unlike Dicer, loss of AGO2 alone did not cause any gross or histological changes in the development of the mouse pancreata. In an effort to understand the underlying mechanisms that are regulated by AGO2, we also studied the activated levels of a variety of signaling molecules in pancreata obtained from different genotypes. Primary analysis indicates that wild type RAS levels are elevated during AGO2 loss and has a bearing on PDAC initiation. Using both Western blot and Immunohistochemical analyses, we demonstrate a previously unknown and critical role for AGO2 in KRAS-MAPK signaling pathway. Citation Format: Sunita Shankar, Jean Tien, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Ingrid A. Apel, Jiaqi Shi, Chandan Kumar-Sinha, Arul M. Chinnaiyan. An in vivo role for Argonaute 2 in KRAS driven pancreatic cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3029. doi:10.1158/1538-7445.AM2017-3029
- Published
- 2017
- Full Text
- View/download PDF
30. Abstract 1362: Argonaute 2 controls RAS activation in mouse embryonic fibroblasts
- Author
-
Malay Mody, Sunita Shankar, Vijaya L. Dommeti, Ronald F. Siebenaler, and Arul M. Chinnaiyan
- Subjects
Cancer Research ,Oncology ,Biology ,Argonaute ,Embryonic stem cell ,Molecular biology ,Cell biology - Abstract
The RAS gene family is among the most commonly mutated genes within cancer, and while much research has elucidated the major downstream pathways, including MAPK and PI3K, little progress has been made in successfully targeting mutant RAS in cancer. We recently identified an interaction between the N terminal domain of Argonaute 2 (AGO2), a core component of RNA-induced silencing complex (RISC), and the Switch II domain of KRAS. Furthermore, this interaction was found in all cell lines tested, expressing either wild-type (WT) or mutant KRAS. We found that stable knockdown of AGO2 in KRAS dependent cell lines lead to a decrease in KRAS protein expression with a subsequent decrease in cellular proliferation. Conversely, the overexpression of AGO2 in these cells lead to both an increase in KRAS expression and oncogenesis. In addition, this interaction inhibits the RNAi function of AGO2 by preventing microRNA unwinding in the presence of oncogenic KRAS compared to WT-KRAS. Despite a clear association between mutant KRAS and AGO2 mediating increased KRAS mediated oncogenesis, the precise function of this interaction remains unclear in normal physiology. In order to better assess the endogenous function of the KRAS-AGO2 interaction, we analyzed two mouse embryonic fibroblast cell lines (NIH 3T3 and MEF) with complete knockout of AGO2. Utilizing a Raf-1 RAS binding domain (RBD) pulldown method, we assessed activated WT-RAS levels in AGO2 null NIH 3T3 and MEF cells. We found that knockout of AGO2 lead to an increase in WT RAS-GTP activation compared to normal control cells. Immunoblot analysis also indicates that AGO2 null fibroblasts lead to increase in RAS downstream signaling through the MAPK/ERK and PI3K/AKT pathways. Furthermore, rescue of AGO2 knockout using full length mouse AGO2 decreased wild type RAS activation and its downstream signaling. Taken together, these observations suggest that the AGO2 interaction may suppress WT-KRAS activation, leading to maintenance of RAS-GDP levels. Using RNA-seq, proteome and microRNA analysis, we have begun to identify the pathways that may be involved in RAS activation in AGO2 null cells. Early analyses indicate that AGO2 controls WT-KRAS levels and activity through multiple mechanisms, laying the foundation for a better understanding of the RAS-AGO2 interaction in normal physiology. Citation Format: Ronald F. Siebenaler, Sunita Shankar, Vijaya L. Dommeti, Malay Mody, Arul Chinnaiyan. Argonaute 2 controls RAS activation in mouse embryonic fibroblasts [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1362. doi:10.1158/1538-7445.AM2017-1362
- Published
- 2017
- Full Text
- View/download PDF
31. Abstract LB-099: BET bromodomain proteins are essential for the oncogenic EWS-fusion driven Ewing Sarcoma
- Author
-
Carl G. Engelke, Zhaohui S. Qin, Irfan A. Asangani, Archana Nallasivam, Tianlei Xu, Vijaya L. Dommeti, Kari Wilder-Romans, Paradesi Naidu Gollavilli, Ingrid A. Apel, Felix Y. Feng, Aishwarya Pawar, and Pranathi Meda Krishnamurthy
- Subjects
Cancer Research ,Oncology ,Cancer research ,medicine ,Sarcoma ,Biology ,medicine.disease ,Virology ,Bromodomain - Abstract
Ewing sarcoma (EWS) is a lethal pediatric small, round, blue cell tumor defined by the characteristic fusion between EWSR1 and a member of the ETS family transcription factor, mostly FLI1 and ERG. Bromodomain and extra-terminal domain (BET) proteins play an essential role in transcription as an epigenetic reader of acetylated histones and transcription factors, and recent studies of inhibitors against these proteins have demonstrated therapeutic potential in various cancers. In this study, we investigated the importance of BET proteins BRD2, BRD3, and BRD4 in EWS-fusion driven transcription program required for maintenance and progression of EWS. Similar to EWS-FLI1 and EWS-ERG, knock-down of BET proteins severely impaired the proliferation and invasion of EWS cells. Gene expression profiling by RNA-Seq showed a common set of genes altered in EWS-FLI1 and BRD2/3/4 knockdown cells implicating a direct role of BET proteins in the transcriptional regulation of EWS-fusion targets. Importantly, BET inhibitor, JQ1, induces apoptosis in EWS cells through an MYC-independent mechanism, and without affecting the EWS-fusion levels. Rather, evidence suggested that BRD4 exists in a complex with EWS-ETS fusion proteins and that its inhibition abrogates the fusion transcriptional program. In vivo, JQ1 inhibited the metastases in chicken CAM model of EWS as well as displayed anti-tumor and anti-metastatic activity in mouse xenograft models of EWS. Together, these results indicate that BET proteins, especially BRD4 interacts with EWS-ETS fusion proteins and this interaction is necessary for the EWS-fusion mediated oncogenesis. Moreover, this study provides a clear rationale for the clinical utility of BET inhibitors in treating metastatic Ewing sarcoma patients. Citation Format: Paradesi N. Gollavilli, Aishwarya Pawar, Kari Wilder-Romans, Carl Engelke, Vijaya L. Dommeti, Pranathi M. Krishnamurthy, Archana Nallasivam, Ingrid A. Apel, Tianlei Xu, Zhaohui S. Qin, Felix Y. Feng, Irfan A. Asangani. BET bromodomain proteins are essential for the oncogenic EWS-fusion driven Ewing Sarcoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr LB-099. doi:10.1158/1538-7445.AM2017-LB-099
- Published
- 2017
- Full Text
- View/download PDF
32. Abstract A05: An in vivo model reveals a role for Argonaute 2 in oncogenic KRAS driven pancreatic cancer initiation
- Author
-
Sunita Shankar, Vijaya L. Dommeti, Arul M. Chinnaiyan, Sylvia Zelenka-Wang, and Jean Tien
- Subjects
Cancer Research ,Cell signaling ,biology ,Argonaute ,medicine.disease_cause ,medicine.disease ,Molecular biology ,Oncology ,Pancreatic cancer ,Cancer cell ,microRNA ,medicine ,Cancer research ,biology.protein ,KRAS ,Carcinogenesis ,Dicer - Abstract
Oncogenic mutations in RAS provide a compelling yet intractable therapeutic target. Using co-immunoprecipitation mass spectrometry, we recently identified an interaction between RAS and Argonaute 2 (AGO2), the core component of the RNA silencing machinery exerting translational control of mRNA transcripts. RAS and AGO2 co-sediment and co-localize in intracellular endomembrane bound organelles. The AGO2 N-terminal domain directly binds the KRAS Switch II region, irrespective of GDP/GTP bound to RAS. Functionally, we observed that AGO2 was required for maximal oncogenic KRAS levels and AGO2 knock-down attenuates cell proliferation in mutant KRAS-dependent cells. Intriguingly, AGO2 mediated microRNA processing (RISC activity) was attenuated in cancer cells expressing mutant KRAS compared to those expressing wild type RAS. Yet, these initial investigations using cell line models do not reveal a clear role for AGO2 in KRAS driven oncogenesis. Therefore, we probed the role of AGO2 in the well-established pancreatic ductal adenocarcinoma (PDAC) mouse model driven by oncogenic KRAS. Towards this end, we generated LSL-KrasG12D; Pft1a Cre; AGO2flox/flox (KCA) mice. Preliminary analysis (8-10 weeks of age) suggests that compared to LSL-KRASG12D (KC) mice, homozygous loss of AGO2 results in increased KRAS driven Acinar to Ductal Metaplasia (ADM) phenotype considered as precursors of PanIN (pancreatic intraepithelial neoplasia) and PDAC. This mirrors the phenotype of loss of Dicer, another microRNA processing enzyme, in the same model further reinforcing a role for microRNAs in restraining KRAS oncogenic programs. Yet unlike Dicer, loss of AGO2 alone did not cause any gross or histological changes in the development of the mouse pancreata. In an effort to understand the underlying mechanisms that are regulated by AGO2, we also studied the activated levels of a variety of signaling molecules in pancreata obtained from different genotypes. Using both Western blot and Immunohistochemical analyses, we demonstrate a previously unknown and critical role for AGO2 in KRAS-MAPK signaling pathway. Together our data from the in vivo mouse model suggests that AGO2 is an important regulator of KRAS signaling and oncogenic KRAS alters the translational machinery through its interaction with AGO2. Citation Format: Sunita Shankar, Jean Tien, Vijaya L. Dommeti, Sylvia Zelenka-Wang, Arul M. Chinnaiyan. An in vivo model reveals a role for Argonaute 2 in oncogenic KRAS driven pancreatic cancer initiation. [abstract]. In: Proceedings of the AACR Special Conference on Translational Control of Cancer: A New Frontier in Cancer Biology and Therapy; 2016 Oct 27-30; San Francisco, CA. Philadelphia (PA): AACR; Cancer Res 2017;77(6 Suppl):Abstract nr A05.
- Published
- 2017
- Full Text
- View/download PDF
33. Therapeutic targeting of BET bromodomain proteins in castration-resistant prostate cancer
- Author
-
Shaomeng Wang, Xiaoju Wang, June Escara-Wilke, Vijaya L. Dommeti, Sudheer Dhanireddy, Mathew K. Iyer, Irfan A. Asangani, Kari Wilder-Romans, Rendong Yang, Felix Y. Feng, Rohit Malik, Carl G. Engelke, Yi-Mi Wu, Arul M. Chinnaiyan, Xiaojun Jing, Zhaohui S. Qin, Marcin Cieslik, and Xuhong Cao
- Subjects
Male ,BRD4 ,Invited Research Highlight ,Oncogene Proteins, Fusion ,Cell Cycle Proteins ,Biology ,urologic and male genital diseases ,Article ,Epigenesis, Genetic ,Fusion gene ,BET inhibitor ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Enzalutamide ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Cancer ,Nuclear Proteins ,Androgen Antagonists ,Azepines ,Triazoles ,medicine.disease ,3. Good health ,Bromodomain ,Protein Structure, Tertiary ,Androgen receptor ,Disease Models, Animal ,Prostatic Neoplasms, Castration-Resistant ,chemistry ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Androgens ,Signal Transduction ,Transcription Factors - Abstract
Men who develop metastatic castration-resistant prostate cancer (CRPC) invariably succumb to the disease. Progression to CRPC after androgen ablation therapy is predominantly driven by deregulated androgen receptor (AR) signalling. Despite the success of recently approved therapies targeting AR signalling, such as abiraterone and second-generation anti-androgens including MDV3100 (also known as enzalutamide), durable responses are limited, presumably owing to acquired resistance. Recently, JQ1 and I-BET762 two selective small-molecule inhibitors that target the amino-terminal bromodomains of BRD4, have been shown to exhibit anti-proliferative effects in a range of malignancies. Here we show that AR-signalling-competent human CRPC cell lines are preferentially sensitive to bromodomain and extraterminal (BET) inhibition. BRD4 physically interacts with the N-terminal domain of AR and can be disrupted by JQ1 (refs 11, 13). Like the direct AR antagonist MDV3100, JQ1 disrupted AR recruitment to target gene loci. By contrast with MDV3100, JQ1 functions downstream of AR, and more potently abrogated BRD4 localization to AR target loci and AR-mediated gene transcription, including induction of the TMPRSS2-ERG gene fusion and its oncogenic activity. In vivo, BET bromodomain inhibition was more efficacious than direct AR antagonism in CRPC xenograft mouse models. Taken together, these studies provide a novel epigenetic approach for the concerted blockade of oncogenic drivers in advanced prostate cancer.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.