33 results on '"Emanuela Dylgjeri"'
Search Results
2. PARP‐1 regulates DNA repair factor availability
- Author
-
Matthew J Schiewer, Amy C Mandigo, Nicolas Gordon, Fangjin Huang, Sanchaika Gaur, Renée de Leeuw, Shuang G Zhao, Joseph Evans, Sumin Han, Theodore Parsons, Ruth Birbe, Peter McCue, Christopher McNair, Saswati N Chand, Ylenia Cendon‐Florez, Peter Gallagher, Jennifer J McCann, Neermala Poudel Neupane, Ayesha A Shafi, Emanuela Dylgjeri, Lucas J Brand, Tapio Visakorpi, Ganesh V Raj, Costas D Lallas, Edouard J Trabulsi, Leonard G Gomella, Adam P Dicker, Wm. Kevin Kelly, Benjamin E Leiby, Beatrice Knudsen, Felix Y Feng, and Karen E Knudsen
- Subjects
DNA repair ,E2F1 ,PARP ,transcription ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract PARP‐1 holds major functions on chromatin, DNA damage repair and transcriptional regulation, both of which are relevant in the context of cancer. Here, unbiased transcriptional profiling revealed the downstream transcriptional profile of PARP‐1 enzymatic activity. Further investigation of the PARP‐1‐regulated transcriptome and secondary strategies for assessing PARP‐1 activity in patient tissues revealed that PARP‐1 activity was unexpectedly enriched as a function of disease progression and was associated with poor outcome independent of DNA double‐strand breaks, suggesting that enhanced PARP‐1 activity may promote aggressive phenotypes. Mechanistic investigation revealed that active PARP‐1 served to enhance E2F1 transcription factor activity, and specifically promoted E2F1‐mediated induction of DNA repair factors involved in homologous recombination (HR). Conversely, PARP‐1 inhibition reduced HR factor availability and thus acted to induce or enhance “BRCA‐ness”. These observations bring new understanding of PARP‐1 function in cancer and have significant ramifications on predicting PARP‐1 inhibitor function in the clinical setting.
- Published
- 2018
- Full Text
- View/download PDF
3. Figure 1 from Novel Oncogenic Transcription Factor Cooperation in RB-Deficient Cancer
- Author
-
Karen E. Knudsen, Johann S. de Bono, Chris M. McNair, Matthew J. Schiewer, Irina A. Vasilevskaya, Galina Semenova, Emanuela Dylgjeri, Lewis Gallagher, Denisa Bogdan, Talya S. Laufer, Wei Yuan, Jennifer J. McCann, Ayesha A. Shafi, and Amy C. Mandigo
- Abstract
Supplemental Figure 1
- Published
- 2023
4. Figure 3 from Novel Oncogenic Transcription Factor Cooperation in RB-Deficient Cancer
- Author
-
Karen E. Knudsen, Johann S. de Bono, Chris M. McNair, Matthew J. Schiewer, Irina A. Vasilevskaya, Galina Semenova, Emanuela Dylgjeri, Lewis Gallagher, Denisa Bogdan, Talya S. Laufer, Wei Yuan, Jennifer J. McCann, Ayesha A. Shafi, and Amy C. Mandigo
- Abstract
Supplemental Figure 3
- Published
- 2023
5. Supplementary Figure Legends from Novel Oncogenic Transcription Factor Cooperation in RB-Deficient Cancer
- Author
-
Karen E. Knudsen, Johann S. de Bono, Chris M. McNair, Matthew J. Schiewer, Irina A. Vasilevskaya, Galina Semenova, Emanuela Dylgjeri, Lewis Gallagher, Denisa Bogdan, Talya S. Laufer, Wei Yuan, Jennifer J. McCann, Ayesha A. Shafi, and Amy C. Mandigo
- Abstract
Supplemental Figure Legends
- Published
- 2023
6. Figure 4 from Novel Oncogenic Transcription Factor Cooperation in RB-Deficient Cancer
- Author
-
Karen E. Knudsen, Johann S. de Bono, Chris M. McNair, Matthew J. Schiewer, Irina A. Vasilevskaya, Galina Semenova, Emanuela Dylgjeri, Lewis Gallagher, Denisa Bogdan, Talya S. Laufer, Wei Yuan, Jennifer J. McCann, Ayesha A. Shafi, and Amy C. Mandigo
- Abstract
Supplemental Figure 4
- Published
- 2023
7. Data from Novel Oncogenic Transcription Factor Cooperation in RB-Deficient Cancer
- Author
-
Karen E. Knudsen, Johann S. de Bono, Chris M. McNair, Matthew J. Schiewer, Irina A. Vasilevskaya, Galina Semenova, Emanuela Dylgjeri, Lewis Gallagher, Denisa Bogdan, Talya S. Laufer, Wei Yuan, Jennifer J. McCann, Ayesha A. Shafi, and Amy C. Mandigo
- Abstract
The retinoblastoma tumor suppressor (RB) is a critical regulator of E2F-dependent transcription, controlling a multitude of protumorigenic networks including but not limited to cell-cycle control. Here, genome-wide assessment of E2F1 function after RB loss in isogenic models of prostate cancer revealed unexpected repositioning and cooperation with oncogenic transcription factors, including the major driver of disease progression, the androgen receptor (AR). Further investigation revealed that observed AR/E2F1 cooperation elicited novel transcriptional networks that promote cancer phenotypes, especially as related to evasion of cell death. These observations were reflected in assessment of human disease, indicating the clinical relevance of the AR/E2F1 cooperome in prostate cancer. Together, these studies reveal new mechanisms by which RB loss induces cancer progression and highlight the importance of understanding the targets of E2F1 function.Significance:This study identifies that RB loss in prostate cancer drives cooperation between AR and E2F1 as coregulators of transcription, which is linked to the progression of advanced disease.
- Published
- 2023
8. Supplementary Tables from Pleiotropic Impact of DNA-PK in Cancer and Implications for Therapeutic Strategies
- Author
-
Karen E. Knudsen, Dana Rathkopf, Kristin Hege, Ellen H. Filvaroff, Felix Y. Feng, Edouard J. Trabulsi, Costas D. Lallas, Leonard G. Gomella, Talya S. Laufer, Nicolas Gordon, Irina Vasilevskaya, Lucas J. Brand, Matthew J. Schiewer, Saswati N. Chand, Amy C. Mandigo, Jennifer J. McCann, Vishal Kothari, Renée de Leeuw, Benjamin E. Leiby, Ayesha A. Shafi, Peter A. McCue, Heather K. Raymon, Jonathan F. Goodwin, Christopher McNair, and Emanuela Dylgjeri
- Abstract
Table S1, Primer sequences used for gene expression analysis. Table S2, PDE pathological characteristics.
- Published
- 2023
9. Supplementary Data from USP22 Functions as an Oncogenic Driver in Prostate Cancer by Regulating Cell Proliferation and DNA Repair
- Author
-
Karen E. Knudsen, William F. Ostrander, Lisa D. Berman-Booty, Steven B. McMahon, Timothy J. Stanek, Peter Gallagher, Randy S. Schrecengost, Matthew J. Schiewer, Amy C. Mandigo, Emanuela Dylgjeri, Christopher McNair, Ayesha A. Shafi, Neermala Poudel Neupane, Irina A. Vasilevskaya, and Jennifer J. McCann
- Abstract
SF1: Analysis of USP22 expression with known drivers of disease progression; SF2: USP22 modulates DNA repair factors expression and survival after DNA damage; SF3: Genetically engineered mouse model of tumor-associated USP22 expression; SF4: The USP22-sensitive ubiquitylome reveals altered modification of DNA repair proteins; SF5: USP22 deubiquitylates the NER protein XPC, modulating foci formation.
- Published
- 2023
10. Supplementary Data from Pleiotropic Impact of DNA-PK in Cancer and Implications for Therapeutic Strategies
- Author
-
Karen E. Knudsen, Dana Rathkopf, Kristin Hege, Ellen H. Filvaroff, Felix Y. Feng, Edouard J. Trabulsi, Costas D. Lallas, Leonard G. Gomella, Talya S. Laufer, Nicolas Gordon, Irina Vasilevskaya, Lucas J. Brand, Matthew J. Schiewer, Saswati N. Chand, Amy C. Mandigo, Jennifer J. McCann, Vishal Kothari, Renée de Leeuw, Benjamin E. Leiby, Ayesha A. Shafi, Peter A. McCue, Heather K. Raymon, Jonathan F. Goodwin, Christopher McNair, and Emanuela Dylgjeri
- Abstract
Supplementary Figures 1-6
- Published
- 2023
11. Data from Pleiotropic Impact of DNA-PK in Cancer and Implications for Therapeutic Strategies
- Author
-
Karen E. Knudsen, Dana Rathkopf, Kristin Hege, Ellen H. Filvaroff, Felix Y. Feng, Edouard J. Trabulsi, Costas D. Lallas, Leonard G. Gomella, Talya S. Laufer, Nicolas Gordon, Irina Vasilevskaya, Lucas J. Brand, Matthew J. Schiewer, Saswati N. Chand, Amy C. Mandigo, Jennifer J. McCann, Vishal Kothari, Renée de Leeuw, Benjamin E. Leiby, Ayesha A. Shafi, Peter A. McCue, Heather K. Raymon, Jonathan F. Goodwin, Christopher McNair, and Emanuela Dylgjeri
- Abstract
Purpose:DNA-dependent protein kinase catalytic subunit (DNA-PK) is a pleiotropic kinase involved in DNA repair and transcriptional regulation. DNA-PK is deregulated in selected cancer types and is strongly associated with poor outcome. The underlying mechanisms by which DNA-PK promotes aggressive tumor phenotypes are not well understood. Here, unbiased molecular investigation in clinically relevant tumor models reveals novel functions of DNA-PK in cancer.Experimental Design: DNA-PK function was modulated using both genetic and pharmacologic methods in a series of in vitro models, in vivo xenografts, and patient-derived explants (PDE), and the impact on the downstream signaling and cellular cancer phenotypes was discerned. Data obtained were used to develop novel strategies for combinatorial targeting of DNA-PK and hormone signaling pathways.Results:Key findings reveal that (i) DNA-PK regulates tumor cell proliferation; (ii) pharmacologic targeting of DNA-PK suppresses tumor growth both in vitro, in vivo, and ex vivo; (iii) DNA-PK transcriptionally regulates the known DNA-PK–mediated functions as well as novel cancer-related pathways that promote tumor growth; (iv) dual targeting of DNA-PK/TOR kinase (TORK) transcriptionally upregulates androgen signaling, which can be mitigated using the androgen receptor (AR) antagonist enzalutamide; (v) cotargeting AR and DNA-PK/TORK leads to the expansion of antitumor effects, uncovering the modulation of novel, highly relevant protumorigenic cancer pathways; and (viii) cotargeting DNA-PK/TORK and AR has cooperative growth inhibitory effects in vitro and in vivo.Conclusions:These findings uncovered novel DNA-PK transcriptional regulatory functions and led to the development of a combinatorial therapeutic strategy for patients with advanced prostate cancer, currently being tested in the clinical setting.
- Published
- 2023
12. Supplementary Figure from A Novel Role for DNA-PK in Metabolism by Regulating Glycolysis in Castration-Resistant Prostate Cancer
- Author
-
Karen E. Knudsen, William K. Kelly, Jeff Holst, Lisa M. Butler, Jason S. Carroll, Erin L. Seifert, Leonard G. Gomella, Peter A. McCue, Costas D. Lallas, Matthew J. Schiewer, Irina Vasilevskaya, Christopher M. McNair, Saswati Chand, Amy C. Mandigo, Jennifer J. McCann, Swati Irani, Jonathan F. Goodwin, Angel Pang, Yi F. Guan, Peter T. Gallagher, Galina Semenova, Ayesha A. Shafi, Vishal Kothari, and Emanuela Dylgjeri
- Abstract
Supplementary Figure from A Novel Role for DNA-PK in Metabolism by Regulating Glycolysis in Castration-Resistant Prostate Cancer
- Published
- 2023
13. Data from A Novel Role for DNA-PK in Metabolism by Regulating Glycolysis in Castration-Resistant Prostate Cancer
- Author
-
Karen E. Knudsen, William K. Kelly, Jeff Holst, Lisa M. Butler, Jason S. Carroll, Erin L. Seifert, Leonard G. Gomella, Peter A. McCue, Costas D. Lallas, Matthew J. Schiewer, Irina Vasilevskaya, Christopher M. McNair, Saswati Chand, Amy C. Mandigo, Jennifer J. McCann, Swati Irani, Jonathan F. Goodwin, Angel Pang, Yi F. Guan, Peter T. Gallagher, Galina Semenova, Ayesha A. Shafi, Vishal Kothari, and Emanuela Dylgjeri
- Abstract
Purpose:DNA-dependent protein kinase catalytic subunit (DNA-PKcs, herein referred as DNA-PK) is a multifunctional kinase of high cancer relevance. DNA-PK is deregulated in multiple tumor types, including prostate cancer, and is associated with poor outcomes. DNA-PK was previously nominated as a therapeutic target and DNA-PK inhibitors are currently undergoing clinical investigation. Although DNA-PK is well studied in DNA repair and transcriptional regulation, much remains to be understood about the way by which DNA-PK drives aggressive disease phenotypes.Experimental Design:Here, unbiased proteomic and metabolomic approaches in clinically relevant tumor models uncovered a novel role of DNA-PK in metabolic regulation of cancer progression. DNA-PK regulation of metabolism was interrogated using pharmacologic and genetic perturbation using in vitro cell models, in vivo xenografts, and ex vivo in patient-derived explants (PDE).Results:Key findings reveal: (i) the first-in-field DNA-PK protein interactome; (ii) numerous DNA-PK novel partners involved in glycolysis; (iii) DNA-PK interacts with, phosphorylates (in vitro), and increases the enzymatic activity of glycolytic enzymes ALDOA and PKM2; (iv) DNA-PK drives synthesis of glucose-derived pyruvate and lactate; (v) DNA-PK regulates glycolysis in vitro, in vivo, and ex vivo; and (vi) combination of DNA-PK inhibitor with glycolytic inhibitor 2-deoxyglucose leads to additive anti-proliferative effects in aggressive disease.Conclusions:Findings herein unveil novel DNA-PK partners, substrates, and function in prostate cancer. DNA-PK impacts glycolysis through direct interaction with glycolytic enzymes and modulation of enzymatic activity. These events support energy production that may contribute to generation and/or maintenance of DNA-PK–mediated aggressive disease phenotypes.
- Published
- 2023
14. Data from USP22 Functions as an Oncogenic Driver in Prostate Cancer by Regulating Cell Proliferation and DNA Repair
- Author
-
Karen E. Knudsen, William F. Ostrander, Lisa D. Berman-Booty, Steven B. McMahon, Timothy J. Stanek, Peter Gallagher, Randy S. Schrecengost, Matthew J. Schiewer, Amy C. Mandigo, Emanuela Dylgjeri, Christopher McNair, Ayesha A. Shafi, Neermala Poudel Neupane, Irina A. Vasilevskaya, and Jennifer J. McCann
- Abstract
Emerging evidence indicates the deubiquitinase USP22 regulates transcriptional activation and modification of target substrates to promote pro-oncogenic phenotypes. Here, in vivo characterization of tumor-associated USP22 upregulation and unbiased interrogation of USP22-regulated functions in vitro demonstrated critical roles for USP22 in prostate cancer. Specifically, clinical datasets validated that USP22 expression is elevated in prostate cancer, and a novel murine model demonstrated a hyperproliferative phenotype with prostate-specific USP22 overexpression. Accordingly, upon overexpression or depletion of USP22, enrichment of cell-cycle and DNA repair pathways was observed in the USP22-sensitive transcriptome and ubiquitylome using prostate cancer models of clinical relevance. Depletion of USP22 sensitized cells to genotoxic insult, and the role of USP22 in response to genotoxic insult was further confirmed using mouse adult fibroblasts from the novel murine model of USP22 expression. As it was hypothesized that USP22 deubiquitylates target substrates to promote protumorigenic phenotypes, analysis of the USP22-sensitive ubiquitylome identified the nucleotide excision repair protein, XPC, as a critical mediator of the USP22-mediated response to genotoxic insult. Thus, XPC undergoes deubiquitylation as a result of USP22 function and promotes USP22-mediated survival to DNA damage. Combined, these findings reveal unexpected functions of USP22 as a driver of protumorigenic phenotypes and have significant implications for the role of USP22 in therapeutic outcomes.Significance:The studies herein present a novel mouse model of tumor-associated USP22 overexpression and implicate USP22 in modulation of cellular survival and DNA repair, in part through regulation of XPC.
- Published
- 2023
15. PARP inhibition and pharmacological ascorbate demonstrate synergy in castration-resistant prostate cancer
- Author
-
Nicolas Gordon, Peter T. Gallagher, Neermala Poudel Neupane, Amy C. Mandigo, Jennifer K. McCann, Emanuela Dylgjeri, Irina Vasilevskaya, Christopher McNair, Channing J. Paller, Wm. Kevin Kelly, Karen E. Knudsen, Ayesha A. Shafi, and Matthew J. Schiewer
- Subjects
Article - Abstract
Prostate cancer (PCa) is the second leading cause of cancer death for men in the United States. While organ-confined disease has reasonable expectation of cure, metastatic PCa is universally fatal upon recurrence during hormone therapy, a stage termed castration-resistant prostate cancer (CRPC). Until such time as molecularly defined subtypes can be identified and targeted using precision medicine, it is necessary to investigate new therapies that may apply to the CRPC population as a whole.The administration of ascorbate, more commonly known as ascorbic acid or Vitamin C, has proved lethal to and highly selective for a variety of cancer cell types. There are several mechanisms currently under investigation to explain how ascorbate exerts anti-cancer effects. A simplified model depicts ascorbate as a pro-drug for reactive oxygen species (ROS), which accumulate intracellularly and generate DNA damage. It was therefore hypothesized that poly(ADP-ribose) polymerase (PARP) inhibitors, by inhibiting DNA damage repair, would augment the toxicity of ascorbate.ResultsTwo distinct CRPC models were found to be sensitive to physiologically relevant doses of ascorbate. Moreover, additional studies indicate that ascorbate inhibits CRPC growthin vitrovia multiple mechanisms including disruption of cellular energy dynamics and accumulation of DNA damage. Combination studies were performed in CRPC models with ascorbate in conjunction with escalating doses of three different PARP inhibitors (niraparib, olaparib, and talazoparib). The addition of ascorbate augmented the toxicity of all three PARP inhibitors and proved synergistic with olaparib in both CRPC models. Finally, the combination of olaparib and ascorbate was testedin vivoin both castrated and non-castrated models. In both cohorts, the combination treatment significantly delayed tumor growth compared to monotherapy or untreated control.ConclusionsThese data indicate that pharmacological ascorbate is an effective monotherapy at physiological concentrations and kills CRPC cells. Ascorbate-induced tumor cell death was associated with disruption of cellular energy dynamics and accumulation of DNA damage. The addition of PARP inhibition increased the extent of DNA damage and proved effective at slowing CRPC growth bothin vitroandin vivo. These findings nominate ascorbate and PARPi as a novel therapeutic regimen that has the potential to improve CRPC patient outcomes.
- Published
- 2023
16. A Novel Role for DNA-PK in Metabolism by Regulating Glycolysis in Castration-Resistant Prostate Cancer
- Author
-
Emanuela Dylgjeri, Vishal Kothari, Ayesha A. Shafi, Galina Semenova, Peter T. Gallagher, Yi F. Guan, Angel Pang, Jonathan F. Goodwin, Swati Irani, Jennifer J. McCann, Amy C. Mandigo, Saswati Chand, Christopher M. McNair, Irina Vasilevskaya, Matthew J. Schiewer, Costas D. Lallas, Peter A. McCue, Leonard G. Gomella, Erin L. Seifert, Jason S. Carroll, Lisa M. Butler, Jeff Holst, William K. Kelly, and Karen E. Knudsen
- Subjects
Male ,Proteomics ,Prostatic Neoplasms, Castration-Resistant ,Cancer Research ,Oncology ,Pyruvate Kinase ,Humans ,DNA ,DNA-Activated Protein Kinase ,Glycolysis - Abstract
Purpose: DNA-dependent protein kinase catalytic subunit (DNA-PKcs, herein referred as DNA-PK) is a multifunctional kinase of high cancer relevance. DNA-PK is deregulated in multiple tumor types, including prostate cancer, and is associated with poor outcomes. DNA-PK was previously nominated as a therapeutic target and DNA-PK inhibitors are currently undergoing clinical investigation. Although DNA-PK is well studied in DNA repair and transcriptional regulation, much remains to be understood about the way by which DNA-PK drives aggressive disease phenotypes. Experimental Design: Here, unbiased proteomic and metabolomic approaches in clinically relevant tumor models uncovered a novel role of DNA-PK in metabolic regulation of cancer progression. DNA-PK regulation of metabolism was interrogated using pharmacologic and genetic perturbation using in vitro cell models, in vivo xenografts, and ex vivo in patient-derived explants (PDE). Results: Key findings reveal: (i) the first-in-field DNA-PK protein interactome; (ii) numerous DNA-PK novel partners involved in glycolysis; (iii) DNA-PK interacts with, phosphorylates (in vitro), and increases the enzymatic activity of glycolytic enzymes ALDOA and PKM2; (iv) DNA-PK drives synthesis of glucose-derived pyruvate and lactate; (v) DNA-PK regulates glycolysis in vitro, in vivo, and ex vivo; and (vi) combination of DNA-PK inhibitor with glycolytic inhibitor 2-deoxyglucose leads to additive anti-proliferative effects in aggressive disease. Conclusions: Findings herein unveil novel DNA-PK partners, substrates, and function in prostate cancer. DNA-PK impacts glycolysis through direct interaction with glycolytic enzymes and modulation of enzymatic activity. These events support energy production that may contribute to generation and/or maintenance of DNA-PK–mediated aggressive disease phenotypes.
- Published
- 2022
17. DNA-PKcs: A Targetable Protumorigenic Protein Kinase
- Author
-
Emanuela, Dylgjeri and Karen E, Knudsen
- Subjects
Clinical Trials as Topic ,Cancer Research ,Immunity ,DNA-Activated Protein Kinase ,Substrate Specificity ,Gene Expression Regulation, Neoplastic ,enzymes and coenzymes (carbohydrates) ,Oncology ,Neoplasms ,Protein Biosynthesis ,Tumor Microenvironment ,Animals ,Humans ,biological phenomena, cell phenomena, and immunity ,Energy Metabolism ,Protein Kinase Inhibitors - Abstract
DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a pleiotropic protein kinase that plays critical roles in cellular processes fundamental to cancer. DNA-PKcs expression and activity are frequently deregulated in multiple hematologic and solid tumors and have been tightly linked to poor outcome. Given the potentially influential role of DNA-PKcs in cancer development and progression, therapeutic targeting of this kinase is being tested in preclinical and clinical settings. This review summarizes the latest advances in the field, providing a comprehensive discussion of DNA-PKcs functions in cancer and an update on the clinical assessment of DNA-PK inhibitors in cancer therapy.
- Published
- 2021
18. Mutant p53 elicits context-dependent pro-tumorigenic phenotypes
- Author
-
Ayesha A. Shafi, Peter T. Gallagher, Renée de Leeuw, Jennifer J. McCann, Emanuela Dylgjeri, Christopher McNair, Neermala Poudel Neupane, Amy C. Mandigo, Matthew J. Schiewer, Irina A. Vasilevskaya, and Karen E. Knudsen
- Subjects
Male ,Cancer Research ,Mutation ,Prostate cancer ,Tumor suppressor gene ,Carcinogenesis ,Mutant ,Cancer ,Prostatic Neoplasms ,DNA-binding domain ,Biology ,medicine.disease ,medicine.disease_cause ,Phenotype ,Article ,Cell biology ,Loss of heterozygosity ,Genetics ,medicine ,Humans ,Tumor Suppressor Protein p53 ,Molecular Biology ,Gene ,Cancer genetics - Abstract
The tumor suppressor gene TP53 is the most frequently mutated gene in numerous cancer types, including prostate cancer (PCa). Specifically, missense mutations in TP53 are selectively enriched in PCa, and cluster to particular “hot spots” in the p53 DNA binding domain with mutation at the R273 residue occurring most frequently. While this residue is similarly mutated to R273C-p53 or R273H-p53 in all cancer types examined, in PCa selective enrichment of R273C-p53 is observed. Importantly, examination of clinical datasets indicated that TP53 heterozygosity can either be maintained or loss of heterozygosity (LOH) occurs. Thus, to mimic tumor-associated mutant p53, R273C-p53 and R273H-p53 isogenic PCa models were developed in the presence or absence of wild-type p53. In the absence of wild-type p53, both R273C-p53 and R273H-p53 exhibited similar loss of DNA binding, transcriptional profiles, and loss of canonical tumor suppressor functions associated with wild-type p53. In the presence of wild-type p53 expression, both R273C-p53 and R273H-p53 supported canonical p53 target gene expression yet elicited distinct cistromic and transcriptional profiles when compared to each other. Moreover, heterozygous modeling of R273C-p53 or R273H-p53 expression resulted in distinct phenotypic outcomes in vitro and in vivo. Thus, mutant p53 acts in a context-dependent manner to elicit pro-tumorigenic transcriptional profiles, providing critical insight into mutant p53-mediated prostate cancer progression.
- Published
- 2021
19. Novel Oncogenic Transcription Factor Cooperation in RB-Deficient Cancer
- Author
-
Galina Semenova, Irina A. Vasilevskaya, Jennifer J. McCann, Matthew J. Schiewer, Christopher McNair, Wei Yuan, Karen E. Knudsen, Emanuela Dylgjeri, Ayesha A. Shafi, Lewis Gallagher, Denisa Bogdan, Amy C. Mandigo, Johann S. de Bono, and Talya S. Laufer
- Subjects
Male ,Cancer Research ,Carcinogenesis ,Cell Survival ,Ubiquitin-Protein Ligases ,Regulator ,Apoptosis ,Biology ,Transfection ,law.invention ,Cohort Studies ,Prostate cancer ,law ,Cell Line, Tumor ,medicine ,Humans ,E2F1 ,Transcription factor ,Oncogene Proteins ,Binding Sites ,Retinoblastoma ,Prostatic Neoplasms ,Cancer ,Oncogenes ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Androgen receptor ,Retinoblastoma Binding Proteins ,Oncology ,Receptors, Androgen ,Gene Knockdown Techniques ,Cancer research ,Suppressor ,biological phenomena, cell phenomena, and immunity ,E2F1 Transcription Factor ,Protein Binding ,Signal Transduction - Abstract
The retinoblastoma tumor suppressor (RB) is a critical regulator of E2F-dependent transcription, controlling a multitude of protumorigenic networks including but not limited to cell-cycle control. Here, genome-wide assessment of E2F1 function after RB loss in isogenic models of prostate cancer revealed unexpected repositioning and cooperation with oncogenic transcription factors, including the major driver of disease progression, the androgen receptor (AR). Further investigation revealed that observed AR/E2F1 cooperation elicited novel transcriptional networks that promote cancer phenotypes, especially as related to evasion of cell death. These observations were reflected in assessment of human disease, indicating the clinical relevance of the AR/E2F1 cooperome in prostate cancer. Together, these studies reveal new mechanisms by which RB loss induces cancer progression and highlight the importance of understanding the targets of E2F1 function. Significance: This study identifies that RB loss in prostate cancer drives cooperation between AR and E2F1 as coregulators of transcription, which is linked to the progression of advanced disease.
- Published
- 2021
20. RB/E2F1 as a Master Regulator of Cancer Cell Metabolism in Advanced Disease
- Author
-
Beshara Sheehan, Josep Domingo-Domenech, Amy C. Mandigo, Alec Paschalis, Wei Yuan, Talya S. Laufer, Yi Fang Guan, Denisa Bogdan, Jennifer J. McCann, Robert B. Den, Christopher McNair, Angel Pang, Peter A. McCue, Johann S. de Bono, Jeff Holst, Karen E. Knudsen, Kexin Xu, Nicolas Gordon, Ayesha A. Shafi, Emanuela Dylgjeri, Peter Gallagher, Chellappagounder Thangavel, Matthew J. Schiewer, Saswati N. Chand, and Irina A. Vasilevskaya
- Subjects
Retinal Neoplasms ,Biology ,Retinoblastoma Protein ,Article ,law.invention ,Transcriptome ,Mice ,law ,Cell Line, Tumor ,medicine ,Animals ,Humans ,E2F1 ,Neoplasm Metastasis ,Retinoblastoma ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Oncology ,Cistrome ,Cancer cell ,Cancer research ,Suppressor ,Reprogramming ,E2F1 Transcription Factor ,Signal Transduction - Abstract
Loss of the retinoblastoma (RB) tumor suppressor protein is a critical step in reprogramming biological networks that drive cancer progression, although mechanistic insight has been largely limited to the impact of RB loss on cell-cycle regulation. Here, isogenic modeling of RB loss identified disease stage–specific rewiring of E2F1 function, providing the first-in-field mapping of the E2F1 cistrome and transcriptome after RB loss across disease progression. Biochemical and functional assessment using both in vitro and in vivo models identified an unexpected, prominent role for E2F1 in regulation of redox metabolism after RB loss, driving an increase in the synthesis of the antioxidant glutathione, specific to advanced disease. These E2F1-dependent events resulted in protection from reactive oxygen species in response to therapeutic intervention. On balance, these findings reveal novel pathways through which RB loss promotes cancer progression and highlight potentially new nodes of intervention for treating RB-deficient cancers. Significance: This study identifies stage-specific consequences of RB loss across cancer progression that have a direct impact on tumor response to clinically utilized therapeutics. The study herein is the first to investigate the effect of RB loss on global metabolic regulation and link RB/E2F1 to redox control in multiple advanced diseases. This article is highlighted in the In This Issue feature, p. 2113
- Published
- 2021
21. DNA-Dependent Protein Kinase Drives Prostate Cancer Progression through Transcriptional Regulation of the Wnt Signaling Pathway
- Author
-
S. Laura Chang, Jonathan Chou, Edward M. Schaeffer, Shuang G. Zhao, Kari Wilder-Romans, Karen E. Knudsen, David A. Quigley, Housheng Hansen He, Emanuela Dylgjeri, Scott A. Tomlins, Kristina Gabbara, Corey Speers, Dana E. Rathkopf, Jonathan F. Goodwin, Vishal Kothari, Ellen Filvaroff, Justin M. Drake, Yi Yin, Luke A. Gilbert, Arul M. Chinnaiyan, Felix Y. Feng, Alan Ashworth, Kristen Hege, Joseph R. Evans, R. Jeffrey Karnes, Rohit Mehra, Ganesh V. Raj, Daniel E. Spratt, and G. Sun
- Subjects
Male ,0301 basic medicine ,Aging ,Cancer Research ,Transcription, Genetic ,DNA-Activated Protein Kinase ,Mice ,Prostate cancer ,0302 clinical medicine ,Cell Movement ,Transcriptional regulation ,2.1 Biological and endogenous factors ,RNA, Small Interfering ,Neoplasm Metastasis ,Aetiology ,Wnt Signaling Pathway ,Cancer ,Regulation of gene expression ,Tumor ,Kinase ,Prostate Cancer ,Wnt signaling pathway ,Gene Expression Regulation, Neoplastic ,Phenotype ,Oncology ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Disease Progression ,Heterografts ,Transcription ,Protein Binding ,Urologic Diseases ,Oncology and Carcinogenesis ,Biology ,Small Interfering ,Article ,Cell Line ,03 medical and health sciences ,Genetic ,Cell Line, Tumor ,Biomarkers, Tumor ,Genetics ,medicine ,Animals ,Humans ,Oncology & Carcinogenesis ,Protein kinase A ,Transcription factor ,Neoplastic ,Animal ,Gene Expression Profiling ,Prostatic Neoplasms ,medicine.disease ,Gene expression profiling ,Disease Models, Animal ,030104 developmental biology ,Gene Expression Regulation ,Disease Models ,Cancer research ,RNA ,Biomarkers - Abstract
Purpose: Protein kinases are known to play a prominent role in oncogenic progression across multiple cancer subtypes, yet their role in prostate cancer progression remains underexplored. The purpose of this study was to identify kinases that drive prostate cancer progression. Experimental Design: To discover kinases that drive prostate cancer progression, we investigated the association between gene expression of all known kinases and long-term clinical outcomes in tumor samples from 545 patients with high-risk disease. We evaluated the impact of genetic and pharmacologic inhibition of the most significant kinase associated with metastatic progression in vitro and in vivo. Results: DNA-dependent protein kinase (DNAPK) was identified as the most significant kinase associated with metastatic progression in high-risk prostate cancer. Inhibition of DNAPK suppressed the growth of both AR-dependent and AR-independent prostate cancer cells. Gene set enrichment analysis nominated Wnt as the top pathway associated with DNAPK. We found that DNAPK interacts with the Wnt transcription factor LEF1 and is critical for LEF1-mediated transcription. Conclusions: Our data show that DNAPK drives prostate cancer progression through transcriptional regulation of Wnt signaling and is an attractive therapeutic target in aggressive prostate cancer.
- Published
- 2019
22. The circadian cryptochrome, CRY1, is a pro-tumorigenic factor that rhythmically modulates DNA repair
- Author
-
Yanyun Zhu, Wilbert Zwart, Nicolas Gordon, Irina A. Vasilevskaya, Michael Brunner, Andre M. Bergman, Felix Y. Feng, Amy C. Mandigo, Matthew J. Schiewer, Mohammed Alshalalfa, Anton Shostak, Peter Gallagher, Ayesha A. Shafi, Jennifer J. McCann, Karen E. Knudsen, Talya S. Laufer, Saswati N. Chand, Emanuela Dylgjeri, Tesa M. Severson, and Christopher McNair
- Subjects
0301 basic medicine ,Male ,Carcinogenesis ,General Physics and Astronomy ,Datasets as Topic ,medicine.disease_cause ,Transcriptome ,0302 clinical medicine ,Transcriptional regulation ,DNA Breaks, Double-Stranded ,Prospective Studies ,RNA-Seq ,Promoter Regions, Genetic ,Regulation of gene expression ,Multidisciplinary ,Prostate cancer ,Prostate ,Middle Aged ,Cell biology ,G2 Phase Cell Cycle Checkpoints ,Gene Expression Regulation, Neoplastic ,Prostatic Neoplasms, Castration-Resistant ,Cistrome ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Androgens ,Disease Progression ,Chromatin Immunoprecipitation Sequencing ,endocrine system ,animal structures ,DNA repair ,DNA damage ,Science ,Double-strand DNA breaks ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Androgen Receptor Antagonists ,Humans ,Homologous recombination ,Transcriptomics ,Steroid hormones ,Aged ,Retrospective Studies ,Prostatectomy ,fungi ,Recombinational DNA Repair ,General Chemistry ,Cryptochromes ,030104 developmental biology ,sense organs ,Neoplasm Grading ,Follow-Up Studies - Abstract
Mechanisms regulating DNA repair processes remain incompletely defined. Here, the circadian factor CRY1, an evolutionally conserved transcriptional coregulator, is identified as a tumor specific regulator of DNA repair. Key findings demonstrate that CRY1 expression is androgen-responsive and associates with poor outcome in prostate cancer. Functional studies and first-in-field mapping of the CRY1 cistrome and transcriptome reveal that CRY1 regulates DNA repair and the G2/M transition. DNA damage stabilizes CRY1 in cancer (in vitro, in vivo, and human tumors ex vivo), which proves critical for efficient DNA repair. Further mechanistic investigation shows that stabilized CRY1 temporally regulates expression of genes required for homologous recombination. Collectively, these findings reveal that CRY1 is hormone-induced in tumors, is further stabilized by genomic insult, and promotes DNA repair and cell survival through temporal transcriptional regulation. These studies identify the circadian factor CRY1 as pro-tumorigenic and nominate CRY1 as a new therapeutic target., Cryptochrome 1 (CRY1) is a transcriptional coregulator associated with the circadian clock. Here the authors reveal that CRY1 is hormone-regulated, stabilized by genomic insult, and promotes DNA repair and cell survival through temporal transcriptional regulation.
- Published
- 2021
23. USP22 functions as an oncogenic driver in prostate cancer by regulating cell proliferation and DNA repair
- Author
-
Karen E. Knudsen, Christopher McNair, Emanuela Dylgjeri, Irina A. Vasilevskaya, William F. Ostrander, Lisa D. Berman-Booty, Timothy J. Stanek, Jennifer J. McCann, Amy C. Mandigo, Neermala Poudel Neupane, Randy S. Schrecengost, Matthew J. Schiewer, Steven B. McMahon, Ayesha A. Shafi, and Peter T. Gallagher
- Subjects
0301 basic medicine ,Male ,Cancer Research ,DNA Repair ,DNA repair ,DNA damage ,Carcinogenesis ,Apoptosis ,Biology ,Article ,Transcriptome ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Animals ,Humans ,Cell Proliferation ,Regulation of gene expression ,Cell growth ,Prostatic Neoplasms ,Cell cycle ,Prognosis ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,030104 developmental biology ,DNA Repair Enzymes ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Ubiquitin Thiolesterase ,Nucleotide excision repair ,DNA Damage - Abstract
Emerging evidence indicates the deubiquitinase USP22 regulates transcriptional activation and modification of target substrates to promote pro-oncogenic phenotypes. Here, in vivo characterization of tumor-associated USP22 upregulation and unbiased interrogation of USP22-regulated functions in vitro demonstrated critical roles for USP22 in prostate cancer. Specifically, clinical datasets validated that USP22 expression is elevated in prostate cancer, and a novel murine model demonstrated a hyperproliferative phenotype with prostate-specific USP22 overexpression. Accordingly, upon overexpression or depletion of USP22, enrichment of cell-cycle and DNA repair pathways was observed in the USP22-sensitive transcriptome and ubiquitylome using prostate cancer models of clinical relevance. Depletion of USP22 sensitized cells to genotoxic insult, and the role of USP22 in response to genotoxic insult was further confirmed using mouse adult fibroblasts from the novel murine model of USP22 expression. As it was hypothesized that USP22 deubiquitylates target substrates to promote protumorigenic phenotypes, analysis of the USP22-sensitive ubiquitylome identified the nucleotide excision repair protein, XPC, as a critical mediator of the USP22-mediated response to genotoxic insult. Thus, XPC undergoes deubiquitylation as a result of USP22 function and promotes USP22-mediated survival to DNA damage. Combined, these findings reveal unexpected functions of USP22 as a driver of protumorigenic phenotypes and have significant implications for the role of USP22 in therapeutic outcomes. Significance: The studies herein present a novel mouse model of tumor-associated USP22 overexpression and implicate USP22 in modulation of cellular survival and DNA repair, in part through regulation of XPC.
- Published
- 2019
24. PARP‐1 regulates DNA repair factor availability
- Author
-
Ylenia Cendon-Florez, Edouard J. Trabulsi, Neermala Poudel Neupane, Benjamin E. Leiby, Felix Y. Feng, Saswati N. Chand, Sumin Han, Matthew J. Schiewer, Sanchaika Gaur, Christopher McNair, Lucas J. Brand, Shuang G. Zhao, Amy C. Mandigo, Emanuela Dylgjeri, Peter A. McCue, Ayesha A. Shafi, Ganesh V. Raj, Renée de Leeuw, Beatrice S. Knudsen, Karen E. Knudsen, Costas D. Lallas, Tapio Visakorpi, Nicolas Gordon, Theodore Parsons, Leonard G. Gomella, Peter Gallagher, Ruth Birbe, Wm. Kevin Kelly, Jennifer J. McCann, Fangjin Huang, Joseph R. Evans, and Adam P. Dicker
- Subjects
Male ,0301 basic medicine ,Medicine (General) ,DNA repair ,Poly ADP ribose polymerase ,Poly (ADP-Ribose) Polymerase-1 ,QH426-470 ,Biology ,Cell Line ,PARP ,Transcriptome ,03 medical and health sciences ,R5-920 ,0302 clinical medicine ,Genetics ,Transcriptional regulation ,Animals ,Humans ,E2F1 ,Homologous Recombination ,Research Articles ,Cancer ,Mice, Inbred BALB C ,Gene Expression Profiling ,Prostatic Neoplasms ,E2F1 Transcription Factor ,Immunohistochemistry ,3. Good health ,Chromatin ,Cell biology ,030104 developmental biology ,Tissue Array Analysis ,030220 oncology & carcinogenesis ,Disease Progression ,Molecular Medicine ,transcription ,Homologous recombination ,Research Article - Abstract
PARP‐1 holds major functions on chromatin, DNA damage repair and transcriptional regulation, both of which are relevant in the context of cancer. Here, unbiased transcriptional profiling revealed the downstream transcriptional profile of PARP‐1 enzymatic activity. Further investigation of the PARP‐1‐regulated transcriptome and secondary strategies for assessing PARP‐1 activity in patient tissues revealed that PARP‐1 activity was unexpectedly enriched as a function of disease progression and was associated with poor outcome independent of DNA double‐strand breaks, suggesting that enhanced PARP‐1 activity may promote aggressive phenotypes. Mechanistic investigation revealed that active PARP‐1 served to enhance E2F1 transcription factor activity, and specifically promoted E2F1‐mediated induction of DNA repair factors involved in homologous recombination (HR). Conversely, PARP‐1 inhibition reduced HR factor availability and thus acted to induce or enhance “BRCA‐ness”. These observations bring new understanding of PARP‐1 function in cancer and have significant ramifications on predicting PARP‐1 inhibitor function in the clinical setting.
- Published
- 2018
25. Pleiotropic Impact of DNA-PK in Cancer and Implications for Therapeutic Strategies
- Author
-
Jennifer J. McCann, Vishal Kothari, Jonathan F. Goodwin, Kristin Hege, Heather Raymon, Edouard J. Trabulsi, Peter A. McCue, Christopher McNair, Irina A. Vasilevskaya, Emanuela Dylgjeri, Nicolas Gordon, Ayesha A. Shafi, Renée de Leeuw, Ellen Filvaroff, Benjamin E. Leiby, Leonard G. Gomella, Karen E. Knudsen, Saswati N. Chand, Costas D. Lallas, Amy C. Mandigo, Dana E. Rathkopf, Lucas J. Brand, Talya S. Laufer, Felix Y. Feng, and Matthew J. Schiewer
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Transcription, Genetic ,Antineoplastic Agents ,DNA-Activated Protein Kinase ,Biology ,Article ,03 medical and health sciences ,Prostate cancer ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Neoplasms ,medicine ,Transcriptional regulation ,Androgen Receptor Antagonists ,Biomarkers, Tumor ,Enzalutamide ,Animals ,Humans ,Molecular Targeted Therapy ,Protein kinase A ,Protein Kinase Inhibitors ,Cell Proliferation ,Kinase ,TOR Serine-Threonine Kinases ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Androgen receptor ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,Oncology ,chemistry ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Cancer research - Abstract
Purpose: DNA-dependent protein kinase catalytic subunit (DNA-PK) is a pleiotropic kinase involved in DNA repair and transcriptional regulation. DNA-PK is deregulated in selected cancer types and is strongly associated with poor outcome. The underlying mechanisms by which DNA-PK promotes aggressive tumor phenotypes are not well understood. Here, unbiased molecular investigation in clinically relevant tumor models reveals novel functions of DNA-PK in cancer. Experimental Design: DNA-PK function was modulated using both genetic and pharmacologic methods in a series of in vitro models, in vivo xenografts, and patient-derived explants (PDE), and the impact on the downstream signaling and cellular cancer phenotypes was discerned. Data obtained were used to develop novel strategies for combinatorial targeting of DNA-PK and hormone signaling pathways. Results: Key findings reveal that (i) DNA-PK regulates tumor cell proliferation; (ii) pharmacologic targeting of DNA-PK suppresses tumor growth both in vitro, in vivo, and ex vivo; (iii) DNA-PK transcriptionally regulates the known DNA-PK–mediated functions as well as novel cancer-related pathways that promote tumor growth; (iv) dual targeting of DNA-PK/TOR kinase (TORK) transcriptionally upregulates androgen signaling, which can be mitigated using the androgen receptor (AR) antagonist enzalutamide; (v) cotargeting AR and DNA-PK/TORK leads to the expansion of antitumor effects, uncovering the modulation of novel, highly relevant protumorigenic cancer pathways; and (viii) cotargeting DNA-PK/TORK and AR has cooperative growth inhibitory effects in vitro and in vivo. Conclusions: These findings uncovered novel DNA-PK transcriptional regulatory functions and led to the development of a combinatorial therapeutic strategy for patients with advanced prostate cancer, currently being tested in the clinical setting.
- Published
- 2018
26. Abstract 1851: Novel roles of DNA-PK in metabolic regulation in prostate cancer
- Author
-
Felix Y. Feng, Giorgia Zadra, Ayesha A. Shafi, Vishal Kothari, Jonathan F. Goodwin, Erin L. Seifert, Karen E. Knudsen, and Emanuela Dylgjeri
- Subjects
Cancer Research ,Kinase ,DNA repair ,Cancer ,Biology ,medicine.disease ,Interactome ,DNA-Dependent Protein Kinase Catalytic Subunit ,Metastasis ,Prostate cancer ,Oncology ,Cancer research ,medicine ,Transcriptional regulation - Abstract
DNA dependent protein kinase catalytic subunit (DNA-PK) is a multifunctional kinase involved in repairing double-strand DNA breaks through non-homologous end joining (NHEJ), with parallel roles as a master transcriptional regulator of gene networks supporting cell migration and invasion. In prostate cancer (PCa), DNA-PK was found to be highly upregulated and hyperactivated. Strikingly, DNA-PK is the most deregulated kinase in metastatic castration resistant prostate cancer (CRPC), and is independently predictive of metastasis and overall survival in patients with high-risk disease. Combined, these findings highlight the importance of understanding DNA-PK functions beyond DNA repair and transcriptional regulation, which promote the acquisition of aggressive tumor phenotypes. Rapid immunoprecipitation of endogenous proteins (RIME) was performed to identify the DNA-PK interactome, which uncovered novel DNA-PK interactors that play key roles in metabolism. To assess the role of DNA-PK in regulation of metabolism, metabolic profiles of CRPC models were investigated upon DNA-PK modulation. Data presented will describe novel roles of DNA-PK in cancer metabolism and subsequent effects of DNA-PK modulation in key metabolic pathways. Citation Format: Emanuela Dylgjeri, Jonathan Goodwin, Ayesha Shafi, Vishal Kothari, Giorgia Zadra, Erin Seifert, Felix Feng, Karen Knudsen. Novel roles of DNA-PK in metabolic regulation in prostate cancer [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 1851.
- Published
- 2019
27. Synergistic effects of the PARP inhibitor olaparib and pharmacological ascorbate in castration-resistant prostate cancer
- Author
-
Matthew J. Schiewer, Ayesha A. Shafi, Peter T. Gallagher, Renée de Leeuw, Emanuela Dylgjeri, William Kevin Kelly, Nicolas Gordon, Karen E. Knudsen, Jennifer K. Jones, Amy C. Mandigo, Lucas J. Brand, Neermala Poudel Neupane, and Christopher McNair
- Subjects
Cancer Research ,business.industry ,Castration resistant ,Highly selective ,medicine.disease ,Olaparib ,chemistry.chemical_compound ,Prostate cancer ,Oncology ,chemistry ,PARP inhibitor ,Cancer cell ,Cancer research ,Medicine ,business - Abstract
326 Background: The administration of ascorbate has proved lethal to and highly selective for a variety of cancer cell types; however, despite an increasingly impressive body of evidence, there has not been a robust effort to translate the observed in vitro and in vivo outcomes to the clinic. This is partially due to the fact that the mechanism by which ascorbate exerts its anti-cancer effect is still under investigation. A simplified model depicts ascorbate as a pro-drug for reactive oxygen species (ROS), which accumulate intracellularly and generate DNA damage. It was therefore hypothesized that poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi), by inhibiting DNA damage repair, would augment the toxicity of ascorbate. Methods: In vitro and in vivo models systems queried for anti-tumor effects of PARP inhibitors and ascorbate. Results: Two distinct castration-resistant prostate cancer (CRPC) models were sensitive to ascorbate at physiologically attainable concentrations. These in vitro models were then subjected to treatment with three different PARP inhibitors (olaparib, niraparib, and talazoparib) alone and in combination with ascorbate. The addition of a sub-lethal dose of ascorbate significantly increased cell death across a range of doses for all three PARP inhibitors. A combination index was generated for olaparib and ascorbate in both CRPC models; the results suggest a strongly synergistic relationship between olaparib and ascorbate. Use of a CRPC in vivo model demonstrated that the combination of olaparib and ascorbate significantly increased tumor doubling time compared to vehicle controls and monotherapy. This in vivo efficacy was even more profound in an additional model using castrated mice to mimic the effect of hormone therapy. Additional mechanistic studies are in progress to further investigate the potential for ascorbate and olaparib combination therapy. Conclusions: Ultimately, these data suggest the combination of ascorbate and PARP inhibitors could be an effective treatment for CRPC.
- Published
- 2019
28. WEE1 inhibition in pancreatic cancer cells is dependent on DNA repair status in a context dependent manner
- Author
-
Nicole C. Mambelli-Lisboa, Charles J. Yeo, Jordan M. Winter, Saswati N. Chand, Emanuela Dylgjeri, Mahsa Zarei, Jonathan R. Brody, Michael J. Pishvaian, and Shruti Lal
- Subjects
0301 basic medicine ,DNA Repair ,Organoplatinum Compounds ,DNA damage ,DNA repair ,Mitomycin ,Cell ,Mitosis ,Antineoplastic Agents ,Apoptosis ,Cell Cycle Proteins ,Pyrimidinones ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,Inhibitory Concentration 50 ,0302 clinical medicine ,Cell Line, Tumor ,FANCD2 ,medicine ,Neoplasm ,Humans ,Mutation ,Multidisciplinary ,Nuclear Proteins ,Drug Synergism ,Protein-Tyrosine Kinases ,medicine.disease ,Molecular biology ,3. Good health ,Mitotic inhibitor ,body regions ,Oxaliplatin ,Pancreatic Neoplasms ,030104 developmental biology ,medicine.anatomical_structure ,DNA Repair Enzymes ,Pyrimidines ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Pyrazoles ,Carcinoma, Pancreatic Ductal ,DNA Damage ,Mutagens - Abstract
Pancreatic ductal adenocarcinoma (PDA) is a lethal disease, in part, because of the lack of effective targeted therapeutic options. MK-1775 (also known as AZD1775), a mitotic inhibitor, has been demonstrated to enhance the anti-tumor effects of DNA damaging agents such as gemcitabine. We evaluated the efficacy of MK-1775 alone or in combination with DNA damaging agents (MMC or oxaliplatin) in PDA cell lines that are either DNA repair proficient (DDR-P) or deficient (DDR-D). PDA cell lines PL11, Hs 766T and Capan-1 harboring naturally selected mutations in DNA repair genes FANCC, FANCG and BRCA2 respectively, were less sensitive to MK-1775 as compared to two out of four representative DDR-P (MIA PaCa2 and PANC-1) cell lines. Accordingly, DDR-P cells exhibit reduced sensitivity to MK-1775 upon siRNA silencing of DNA repair genes, BRCA2 or FANCD2, compared to control cells. Only DDR-P cells showed increased apoptosis as a result of early mitotic entry and catastrophe compared to DDR-D cells. Taken together with other recently published reports, our results add another level of evidence that the efficacy of WEE1 inhibition is influenced by the DNA repair status of a cell and may also be dependent on the tumor type and model evaluated.
- Published
- 2016
29. DNA-PKcs-Mediated Transcriptional Regulation Drives Prostate Cancer Progression and Metastasis
- Author
-
Thomas G. Graeber, Justin M. Drake, Jeffry L. Dean, Vishal Kothari, Elai Davicioni, Nicholas A. Graham, Adam E. Snook, Matthew J. Schiewer, Karen E. Knudsen, Jonathan F. Goodwin, Ajay A. Vashisht, James A. Wohlschlegel, Robert B. Den, Scott A. Tomlins, Cory Abate-Shen, Alvaro Aytes, Nima Sharifi, Shuang G. Zhao, Emanuela Dylgjeri, Leonard G. Gomella, R. Jeffrey Karnes, Ruth Birbe, Owen N. Witte, Felix Y. Feng, Jennifer K. Jones, Ziqi Zhu, Christopher McNair, Michael S. Magee, and M. Takhar
- Subjects
Male ,Cancer Research ,DNA-Activated Protein Kinase ,Metastasis ,Androgen ,Prostate cancer ,Mice ,Receptors ,Transcriptional regulation ,2.1 Biological and endogenous factors ,Gene Regulatory Networks ,Aetiology ,DNA-PKcs ,Cancer ,Regulation of gene expression ,Tumor ,Prostate Cancer ,Nuclear Proteins ,Gene Expression Regulation, Neoplastic ,DNA-Binding Proteins ,Oncology ,Receptors, Androgen ,biological phenomena, cell phenomena, and immunity ,Urologic Diseases ,DNA damage ,DNA repair ,Molecular Sequence Data ,Oncology and Carcinogenesis ,Biology ,Article ,Cell Line ,Cell Line, Tumor ,Breast Cancer ,medicine ,Genetics ,Animals ,Humans ,Neoplasm Invasiveness ,Oncology & Carcinogenesis ,Neoplastic ,Neurosciences ,Prostatic Neoplasms ,Cell Biology ,medicine.disease ,enzymes and coenzymes (carbohydrates) ,Gene Expression Regulation ,Tumor progression ,Cancer research ,Neoplasm Transplantation - Abstract
© 2015 Elsevier Inc.. Emerging evidence demonstrates that the DNA repair kinase DNA-PKcs exerts divergent roles in transcriptional regulation of unsolved consequence. Here, invitro and invivo interrogation demonstrate that DNA-PKcs functions as a selective modulator of transcriptional networks that induce cell migration, invasion, and metastasis. Accordingly, suppression of DNA-PKcs inhibits tumor metastases. Clinical assessment revealed that DNA-PKcs is significantly elevated in advanced disease and independently predicts for metastases, recurrence, and reduced overall survival. Further investigation demonstrated that DNA-PKcs in advanced tumors is highly activated, independent of DNA damage indicators. Combined, these findings reveal unexpected DNA-PKcs functions, identify DNA-PKcs as a potent driver of tumor progression and metastases, and nominate DNA-PKcs as a therapeutic target for advanced malignancies. Goodwin etal. identify DNA-PKcs as a promising therapeutic target that drives prostate cancer progression and metastasis through transcriptional regulation. DNA-PKcs is significantly elevated in advanced disease and is an independent predictor of metastasis, recurrence, and poor survival.
- Published
- 2015
30. Abstract LB-264: Preclinical evaluation of DNA-PK as a therapeutic target in prostate cancer
- Author
-
Karen E. Knudsen, Ayesha A. Shafi, Emanuela Dylgjeri, Dana Rathkop, Christopher McNair, Felix Y. Feng, Vishal Kothari, and Jonathan F. Goodwin
- Subjects
Cancer Research ,business.industry ,Kinase ,DNA repair ,Cancer ,medicine.disease ,Metastasis ,Androgen receptor ,Prostate cancer ,chemistry.chemical_compound ,Oncology ,chemistry ,Cancer research ,Medicine ,Enzalutamide ,Protein kinase A ,business - Abstract
Prostatic adenocarcinoma (PCa) is dependent on androgen receptor (AR) signaling at all stages of disease, as AR activation induces both cell proliferation and survival. Though organ–confined disease can be treated, the response is transient. Reactivation of AR leads to castration resistant prostate cancer (CRPC), which remains fatal. Previously published studies demonstrate that AR activation promotes tumor cell survival and proliferation through a feed-forward loop involving the DNA repair factor DNA-dependent protein kinase (DNA-PK). Recent data from our lab has highlighted the role of DNA-PK in DNA damage repair as well as transcriptional regulation of pro-metastatic signaling. Strikingly, DNA-PK is the most deregulated kinase in metastatic CRPC. DNAPK is highly elevated and hyperactivated, and is an independent predictor of metastasis and overall survival in patients with high-risk disease. Concordantly, DNAPK suppression attenuated DNA-PK functions in NHEJ and transcriptional regulation. DNA-PK inhibition is sufficient to prevent proliferation in vitro and ex vivo, as well as tumor metastasis in vivo. Combined these findings highlight importance of DNA-PK functions as a transcriptional regulator and a mediator of NHEJ DNA repair and nominate it as a therapeutic target in PCa. Data presented here will interrogate the translational capacity of a specific DNA-PK inhibitor, a dual DNA-PK/TOR kinase inhibitor and a specific TOR kinase inhibitor in CRPC models. Currently, CC-115, a dual kinase inhibitor, is the only DNA-PK inhibitor in Phase1/2 trial in combination with Enzalutamide in PCa. Unbiased transcriptomic analyses were performed and will be used to identify mechanisms of action that lead to suppression of PCa growth in vitro and ex vivo. These findings will provide insight in the effectiveness of DNA-PK inhibitors as a single agent or combinatorial treatments and provide rational for its placement in the correct clinical space. Citation Format: Emanuela Dylgjeri, Jonathan F. Goodwin, Christopher M. McNair, Ayesha A. Shafi, Vishal Kothari, Felix Feng, Dana Rathkop, Karen Knudsen. Preclinical evaluation of DNA-PK as a therapeutic target in prostate 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 LB-264. doi:10.1158/1538-7445.AM2017-LB-264
- Published
- 2017
31. Abstract 1656: MK-1775 (WEE1 inhibition) lacks efficacy against DNA repair deficient pancreatic cancer cells
- Author
-
Jordan M. Winter, Emanuela Dylgjeri, Charles J. Yeo, Shruti Lal, Saswati N. Chand, and Jonathan R. Brody
- Subjects
Cancer Research ,Programmed cell death ,Pathology ,medicine.medical_specialty ,DNA repair ,DNA damage ,Biology ,Oncology ,Apoptosis ,Cell culture ,Cancer research ,medicine ,Cytotoxic T cell ,Mitosis ,Mitotic catastrophe - Abstract
Introduction: In this study, we explored the possibility of developing a personalized approach of using MK-1775, a potent WEE1 inhibitor, as mono- or combination therapy to treat pancreatic ductal adenocarcinoma (PDA) cells with DNA repair deficiency. Experimental Methods: We treated PDA cells harboring diverse genetic backgrounds with IC50 doses of clinically-relevant DNA damaging agents: mitomycin C (MMC) and the WEE1 inhibitor MK-1775. Drug sensitivity assays were performed in isogenic PDA cells that are either proficient or deficient in Fanconi Anemia (FA)/BRCA2 pathway. In addition, proficient cell lines were treated with siRNA oligos targeted against FANCD2 and BRCA2 genes. Mechanistic studies such as Annexin V staining were performed to measure apoptotic cells upon MMC and MK-1775 treatments. The degree of DNA damage was measured by immunofluorescence (IF) assay using γH2AX antibody. Mitotic entry was analyzed by both IF and flow cytometry analyses using pH3 antibody. Results: Drug sensitivity assays demonstrated that FA/BRCA2-pathway proficient PDA cell lines (MiaPaCa2 and Panc1) are sensitive to the cytotoxic effect of MK-1775 compared to DNA repair-deficient cell lines (Capan1:BRCA2 deficient and Hs766T:FANCG deficient) which showed acute resistance to MK-1775. Immunoblotting showed that MK-1775 efficiently reduces the expression of WEE1 and accordingly phosphorylation of CDK1 in all cell lines. Annexin V staining showed a higher percentage of cell death in the FA/BRCA2-pathway proficient cell lines compared to deficient cell lines when exposed to MK-1775. Furthermore, IF experiments demonstrated that MMC treatment induces γH2AX foci in all cell lines, however, the FA/BRCA2 proficient cell lines showed a higher degree of nuclear abnormality and multi-nucleation after MK-1775 treatment compared to deficient cell lines. In addition, the FA/BRCA2 proficient cell lines showed significantly higher phospho histone 3 (pH3) staining, a marker of mitotic cells. FACS analyses validated that proficient cell lines showed a higher percentage of cells in the mitotic phase when exposed to MK-1775. In addition, we evaluated the cytotoxic effect of MK-1775 in combination with MMC and showed that FA/BRCA2 proficient cells are more sensitive to dual treatment than deficient cells. In addition, immnobloting detected cleaved caspase 3, a marker of apoptosis, after MK-1775 treatment alone or in combination with MMC, demonstrating cells undergoing mitotic catastrophe. We validated the results in the FA/BRCA2-pathway proficient cell lines pre-treated with siFANCD2 or siBRCA2 oligos as compared to the proficient control lines. Conclusions: These results support a paradigm in which identified high risk FA/BRCA2-mutated patients would not benefit from WEE1 inhibitor monotherapy; and thus, would most likely respond better to conventional DNA damaging agent-based therapies (e.g., oxaliplatin or MMC). Citation Format: Shruti Lal, Saswati N. Chand, Emanuela Dylgjeri, Charles J. Yeo, Jordan M. Winter, Jonathan R. Brody. MK-1775 (WEE1 inhibition) lacks efficacy against DNA repair deficient pancreatic cancer cells. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1656. doi:10.1158/1538-7445.AM2015-1656
- Published
- 2015
32. Abstract 1859: Discerning the molecular basis of DNA-PK pro-tumorigenic functions and translational capacity as a therapeutic target in prostate cancer
- Author
-
Jonathan F. Goodwin, Karen E. Knudsen, and Emanuela Dylgjeri
- Subjects
Cancer Research ,DNA repair ,business.industry ,Cancer ,Pharmacology ,medicine.disease ,Transcriptome ,Androgen receptor ,Prostate cancer ,Oncology ,Cistrome ,medicine ,Transcriptional regulation ,Cancer research ,business ,Transcription factor - Abstract
Prostatic adenocarcinoma (PCa) is dependent on androgen receptor (AR) signaling at all stages of disease, as AR activation induces both cell proliferation and survival. Though organ-confined disease can be treated, the response is transient and reactivation of AR results in incurable state of disease (castration resistant prostate cancer, CRPC). Thus, there is a significant need to discern the mechanisms by which aberrant AR activity arises, and to develop new means to clinically target advanced disease. Recently, AR activation was found to promote tumor cell survival and proliferation through a feed-forward loop involving the repair factor DNA-dependent protein kinase (DNA-PK). Emerging new data correlates elevated DNA-PK expression with poor prognosis and identifies DNA-PK as a driver of metastatic signaling. Although the role of DNA-PK activity in DNA repair is well characterized, the underpinning mechanisms and consequences of DNA-PK in transcriptional regulation are understudied. Data to be discussed will build on our new preliminary DNA-PK interactome data to identify sequence-specific transcription factors found in complex with DNA-PK. In order to molecularly dissect DNA-PK transcriptional events, the DNA-PK cistrome and transcriptome will be defined using clinically relevant models, resulting in a detailed understanding of DNA-PK mediated transcriptional regulation. The correlation of high expression of DNA-PK with poor prognosis in advanced PCa, as well as the known roles of DNA-PK in cancer-relevant pathways make DNA-PK a viable therapeutic target for PCa. Despite the current clinical assessment of DNA-PK inhibitors in multiple tumor types, there are no existing trials targeting PCa. Additional studies that assess the efficacy of current DNA-PK inhibitors in PCa will be presented and used to determine the best clinical space for their use. Citation Format: Emanuela Dylgjeri, Jonathan Goodwin, Karen Knudsen. Discerning the molecular basis of DNA-PK pro-tumorigenic functions and translational capacity as a therapeutic target in prostate cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1859. doi:10.1158/1538-7445.AM2015-1859
- Published
- 2015
33. Abstract 1860: DNA-PK-mediated transcriptional regulation drives tumor progression and metastasis
- Author
-
Thomas G. Graeber, Justin M. Drake, Matthew J. Schiewer, Elai Davicioni, Vishal Kothari, James A. Wohlschlegel, Shuang G. Zhao, Mandeep Takhar, Jeffry L. Dean, Christopher McNair, Nima Sharifi, Ziqi Zhu, Emanuela Dylgjeri, Karen E. Knudsen, Robert B. Den, R. Jeffrey Karnes, Owen N. Witte, Michael S. Magee, Scott A. Tomlins, Felix Y. Feng, Jonathan F. Goodwin, Ajay A. Vashisht, and Nicholas A. J. Graham
- Subjects
Cancer Research ,medicine.medical_specialty ,Biology ,medicine.disease ,Metastasis ,chemistry.chemical_compound ,Endocrinology ,Oncology ,chemistry ,Tumor progression ,Internal medicine ,medicine ,Transcriptional regulation ,Cancer research ,DNA - Abstract
Emerging evidence demonstrates that the DNA repair kinase DNA-PK exerts divergent roles in transcriptional regulation of unsolved consequence. Here, in vitro and in vivo interrogation demonstrate that DNA-PK functions as a selective modulator of transcriptional networks that induce cell migration, invasion, and metastasis. Accordingly, suppression of DNA-PK inhibits tumor metastases. Clinical assessment revealed that DNA-PK is significantly elevated in advanced disease, and independently predicts for metastases, recurrence, and reduced overall survival. Further investigation demonstrated that DNA-PK in advanced tumors is highly activated, independently of DNA damage indicators. Combined, these findings put forth new paradigms for DNA-PK function, identify DNA-PK as a potent driver of tumor progression and metastases, and nominate DNA-PK as a therapeutic target for advanced malignancies. Citation Format: Jonathan F. Goodwin, Vishal Kothari, Justin M. Drake, Shuang Zhao, Emanuela Dylgjeri, Jeffry L. Dean, Matthew J. Schiewer, Christopher McNair, Michael S. Magee, Robert B. Den, Ziqi Zhu, Nicholas A. Graham, Ajay A. Vashisht, James A. Wohlschlegel, Thomas G. Graeber, R Jeffrey Karnes, Mandeep Takhar, Elai Davicioni, Scott A. Tomlins, Nima Sharifi, Owen N. Witte, Felix Y. Feng, Karen E. Knudsen. DNA-PK-mediated transcriptional regulation drives tumor progression and metastasis. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1860. doi:10.1158/1538-7445.AM2015-1860
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.