40 results on '"Nora Navone"'
Search Results
2. Supplementary Movie S2 from Critical Role of Endogenous Heme Oxygenase 1 as a Tuner of the Invasive Potential of Prostate Cancer Cells
- Author
-
Elba S. Vazquez, Nora Navone, Roberto Meiss, Belen Elguero, Paola De Luca, Marcelo Salierno, Mercedes Ferrando, Adriana De Siervi, and Geraldine Gueron
- Abstract
Supplementary Movie S2 from Critical Role of Endogenous Heme Oxygenase 1 as a Tuner of the Invasive Potential of Prostate Cancer Cells
- Published
- 2023
3. Supplementary Movie S1 from Critical Role of Endogenous Heme Oxygenase 1 as a Tuner of the Invasive Potential of Prostate Cancer Cells
- Author
-
Elba S. Vazquez, Nora Navone, Roberto Meiss, Belen Elguero, Paola De Luca, Marcelo Salierno, Mercedes Ferrando, Adriana De Siervi, and Geraldine Gueron
- Abstract
Supplementary Movie S1 from Critical Role of Endogenous Heme Oxygenase 1 as a Tuner of the Invasive Potential of Prostate Cancer Cells
- Published
- 2023
4. Data from Critical Role of Endogenous Heme Oxygenase 1 as a Tuner of the Invasive Potential of Prostate Cancer Cells
- Author
-
Elba S. Vazquez, Nora Navone, Roberto Meiss, Belen Elguero, Paola De Luca, Marcelo Salierno, Mercedes Ferrando, Adriana De Siervi, and Geraldine Gueron
- Abstract
Prostate cancer (PCa) is the second leading cause of cancer-associated death in men. Inflammation has been recognized as a risk factor for this disease. Heme oxygenase 1 (HO-1), the inducible isoform of the rate-limiting enzyme in heme degradation, counteracts oxidative and inflammatory damage. Here, we investigated the regulated expression of HO-1 and its functional consequences in PCa. We studied the effect of genetic and pharmacologic disruption of HO-1 in the growth, invasion, and migration in androgen-sensitive (MDA PCa2b and LNCaP) and androgen-insensitive (PC3) PCa cell lines. Our results show that HO-1 levels are markedly decreased in PC3 compared with MDA PCa2b and LNCaP. Hemin treatment increased HO-1 at both protein and mRNA levels in all cell lines and decreased cell proliferation and invasion. Furthermore, overexpression of HO-1 in PC3 resulted in markedly reduced cell proliferation and migration. Accordingly, small interfering RNA–mediated silencing of HO-1 expression in MDA PCa2b cells resulted in increased proliferation and invasion. Using reverse transcription-quantitative PCR–generated gene array, a set of inflammatory and angiogenic genes were upregulated or downregulated in response to HO-1 overexpression identifying matrix metalloprotease 9 (MMP9) as a novel downstream target of HO-1. MMP9 production and activity was downregulated by HO-1 overexpression. Furthermore, PC3 cells stably transfected with HO-1 (PC3HO-1) and controls were injected into nu/nu mice for analysis of in vivo tumor xenograft phenotype. Tumor growth and MMP9 expression was significantly reduced in PC3HO-1 tumors compared with control xenografts. Taken together, these results implicate HO-1 in PCa cell migration and proliferation suggesting its potential role as a therapeutic target in clinical settings. (Mol Cancer Res 2009;7(11):1745–55)
- Published
- 2023
5. Supplementary Data from Critical Role of Endogenous Heme Oxygenase 1 as a Tuner of the Invasive Potential of Prostate Cancer Cells
- Author
-
Elba S. Vazquez, Nora Navone, Roberto Meiss, Belen Elguero, Paola De Luca, Marcelo Salierno, Mercedes Ferrando, Adriana De Siervi, and Geraldine Gueron
- Abstract
Supplementary Data from Critical Role of Endogenous Heme Oxygenase 1 as a Tuner of the Invasive Potential of Prostate Cancer Cells
- Published
- 2023
6. PD04-02 USING GENOMIC AND TRANSCRIPTOMIC PROPERTIES TO DETERMINE ANDROGEN RESPONSE IN DUCTAL PROSTATE CANCERS AND DETERMINE EFFICACY OF POLY(ADP-RIBOSE) POLYMERASE INHIBITORS WITH ANDROGEN SIGNALLING INHIBITORS THERAPY IN VITRO
- Author
-
Weranja Ranasinghe, Melbourne, Australia Patricia Troncoso, Mitchell Lawrence, Nicholas Choo, Birunthi Niranjan, Melissa Papargiris, Hong Wang, Andrew Ryan, Peter Shepherd, Bradley Broom, Ganiraju Manyam, Renea Taylor, Timothy Thompson, Nora Navone, Gail Risbridger, and Brian Chapin
- Subjects
Urology - Published
- 2023
7. Table S5-S7 from Targeting the MYCN–PARP–DNA Damage Response Pathway in Neuroendocrine Prostate Cancer
- Author
-
Timothy C. Thompson, Patricia Troncoso, Nora Navone, Arul M. Chinnaiyan, Ana M. Aparicio, Christopher J. Logothetis, Paul G. Corn, Jeri Kim, Xuhong Cao, Abul Kalam Azad, Sanghee Park, Guang Yang, Jianxiang Wang, Yang Luan, Dimitrios Korentzelos, Spyridon P. Basourakos, Bradley M. Broom, Likun Li, Wenhui Wu, Bo Liu, and Wei Zhang
- Abstract
Table S5. Primer Sequences for qRT-PCR. Table S6. siRNA Sequence. Table S7. Primer Sequences for ChIP-PCR.
- Published
- 2023
8. Figure S3 from In Vivo Hemin Conditioning Targets the Vascular and Immunologic Compartments and Restrains Prostate Tumor Development
- Author
-
Elba S. Vazquez, Diego J. Laderach, Daniel Compagno, Gabriel A. Rabinovich, Nora Navone, Asif Ahmed, Paula M. Berguer, Emiliano G. Ortiz, Roberto P. Meiss, Geraldine Gueron, Lucas D. Gentilini, and Felipe M. Jaworski
- Abstract
Bioinformatic analysis of human prostate adenocarcinoma microarray datasets. Oncomine was used to browse gene expression microarrays data. Box plots depict prostate statistics derived from Grasso et al. LGALS1, CD34, FLT1, FLT4, VEGFA, VEGFC, VCAM1, CD28, CD80 and CD86 were assessed. Each box plot shows the expression levels for each gene in 1) normal prostate gland and 2) prostate adenocarcinoma. * indicates P
- Published
- 2023
9. Table S3 from Targeting the MYCN–PARP–DNA Damage Response Pathway in Neuroendocrine Prostate Cancer
- Author
-
Timothy C. Thompson, Patricia Troncoso, Nora Navone, Arul M. Chinnaiyan, Ana M. Aparicio, Christopher J. Logothetis, Paul G. Corn, Jeri Kim, Xuhong Cao, Abul Kalam Azad, Sanghee Park, Guang Yang, Jianxiang Wang, Yang Luan, Dimitrios Korentzelos, Spyridon P. Basourakos, Bradley M. Broom, Likun Li, Wenhui Wu, Bo Liu, and Wei Zhang
- Abstract
Differential expression results for DDR-M genes in Beltran's and MDA PDX RNAseq datasets.
- Published
- 2023
10. Table S2 from Targeting the MYCN–PARP–DNA Damage Response Pathway in Neuroendocrine Prostate Cancer
- Author
-
Timothy C. Thompson, Patricia Troncoso, Nora Navone, Arul M. Chinnaiyan, Ana M. Aparicio, Christopher J. Logothetis, Paul G. Corn, Jeri Kim, Xuhong Cao, Abul Kalam Azad, Sanghee Park, Guang Yang, Jianxiang Wang, Yang Luan, Dimitrios Korentzelos, Spyridon P. Basourakos, Bradley M. Broom, Likun Li, Wenhui Wu, Bo Liu, and Wei Zhang
- Abstract
Differential expression result for Beltran's 70 classifier genes in PDX RNAseq dataset.
- Published
- 2023
11. Supplementary Data from Radium-223 Treatment Increases Immune Checkpoint Expression in Extracellular Vesicles from the Metastatic Prostate Cancer Bone Microenvironment
- Author
-
Theocharis Panaretakis, Christopher J. Logothetis, Sue-Hwa Lin, Sumit K. Subudhi, Eleni Efstathiou, Gary E. Gallick, Nora Navone, Patricia Troncoso, Marites P. Melancon, Elmer Santos, Guoyu Yu, Song-Chang Lin, Yu-Chen Lee, Leah Guerra, Nila Parikh, Anh Hoang, Namrata Madan, Jian H. Song, Emanuela Gentile, Paul Corn, and Ioulia Vardaki
- Abstract
Supplementary Figures
- Published
- 2023
12. Data from Targeting the MYCN–PARP–DNA Damage Response Pathway in Neuroendocrine Prostate Cancer
- Author
-
Timothy C. Thompson, Patricia Troncoso, Nora Navone, Arul M. Chinnaiyan, Ana M. Aparicio, Christopher J. Logothetis, Paul G. Corn, Jeri Kim, Xuhong Cao, Abul Kalam Azad, Sanghee Park, Guang Yang, Jianxiang Wang, Yang Luan, Dimitrios Korentzelos, Spyridon P. Basourakos, Bradley M. Broom, Likun Li, Wenhui Wu, Bo Liu, and Wei Zhang
- Abstract
Purpose: We investigated MYCN-regulated molecular pathways in castration-resistant prostate cancer (CRPC) classified by morphologic criteria as adenocarcinoma or neuroendocrine to extend the molecular phenotype, establish driver pathways, and identify novel approaches to combination therapy for neuroendocrine prostate cancer (NEPC).Experimental Design and Results: Using comparative bioinformatics analyses of CRPC-Adeno and CRPC-Neuro RNA sequence data from public data sets and a panel of 28 PDX models, we identified a MYCN–PARP–DNA damage response (DDR) pathway that is enriched in CRPC with neuroendocrine differentiation (NED) and CRPC-Neuro. ChIP-PCR assay revealed that N-MYC transcriptionally activates PARP1, PARP2, BRCA1, RMI2, and TOPBP1 through binding to the promoters of these genes. MYCN or PARP1 gene knockdown significantly reduced the expression of MYCN–PARP–DDR pathway genes and NED markers, and inhibition with MYCNsi and/or PARPsi, BRCA1si, or RMI2si significantly suppressed malignant activities, including cell viability, colony formation, and cell migration, in C4-2b4 and NCI-H660 cells. Targeting this pathway with AURKA inhibitor PHA739358 and PARP inhibitor olaparib generated therapeutic effects similar to those of gene knockdown in vitro and significantly suppressed tumor growth in both C4-2b4 and MDACC PDX144-13C subcutaneous models in vivo.Conclusions: Our results identify a novel MYCN–PARP–DDR pathway that is driven by N-MYC in a subset of CRPC-Adeno and in NEPC. Targeting this pathway using in vitro and in vivo CRPC-Adeno and CRPC-Neuro models demonstrated a novel therapeutic strategy for NEPC. Further investigation of N-MYC–regulated DDR gene targets and the biological and clinical significance of MYCN–PARP–DDR signaling will more fully elucidate the importance of the MYCN–PARP–DDR signaling pathway in the development and maintenance of NEPC. Clin Cancer Res; 24(3); 696–707. ©2017 AACR.
- Published
- 2023
13. Data from Radium-223 Treatment Increases Immune Checkpoint Expression in Extracellular Vesicles from the Metastatic Prostate Cancer Bone Microenvironment
- Author
-
Theocharis Panaretakis, Christopher J. Logothetis, Sue-Hwa Lin, Sumit K. Subudhi, Eleni Efstathiou, Gary E. Gallick, Nora Navone, Patricia Troncoso, Marites P. Melancon, Elmer Santos, Guoyu Yu, Song-Chang Lin, Yu-Chen Lee, Leah Guerra, Nila Parikh, Anh Hoang, Namrata Madan, Jian H. Song, Emanuela Gentile, Paul Corn, and Ioulia Vardaki
- Abstract
Purpose:Radium-223 prolongs survival in a fraction of men with bone metastatic prostate cancer (PCa). However, there are no markers for monitoring response and resistance to Radium-223 treatment. Exosomes are mediators of intercellular communication and may reflect response of the bone microenvironment to Radium-223 treatment. We performed molecular profiling of exosomes and compared the molecular profile in patients with favorable and unfavorable overall survival.Experimental Design:We performed exosomal transcriptome analysis in plasma derived from our preclinical models (MDA-PCa 118b tumors, TRAMP-C2/BMP4 PCa) and from the plasma of 25 patients (paired baseline and end of treatment) treated with Radium-223. All samples were run in duplicate, and array data analyzed with fold changes +2 to −2 and P < 0.05.Results:We utilized the preclinical models to establish that genes derived from the tumor and the tumor-associated bone microenvironment (bTME) are differentially enriched in plasma exosomes upon Radium-223 treatment. The mouse transcriptome analysis revealed changes in bone-related and DNA damage repair–related pathways. Similar findings were observed in plasma-derived exosomes from patients treated with Radium-223 detected changes. In addition, exosomal transcripts detected immune-suppressors (e.g., PD-L1) that were associated with shorter survival to Radium-223. Treatment of the Myc-CaP mouse model with a combination of Radium-223 and immune checkpoint therapy (ICT) resulted in greater efficacy than monotherapy.Conclusions:These clinical and coclinical analyses showed that RNA profiling of plasma exosomes may be used for monitoring the bTME in response to treatment and that ICT may be used to increase the efficacy of Radium-223.
- Published
- 2023
14. Data from In Vivo Hemin Conditioning Targets the Vascular and Immunologic Compartments and Restrains Prostate Tumor Development
- Author
-
Elba S. Vazquez, Diego J. Laderach, Daniel Compagno, Gabriel A. Rabinovich, Nora Navone, Asif Ahmed, Paula M. Berguer, Emiliano G. Ortiz, Roberto P. Meiss, Geraldine Gueron, Lucas D. Gentilini, and Felipe M. Jaworski
- Abstract
Purpose: Conditioning strategies constitute a relatively unexplored and exciting opportunity to shape tumor fate by targeting the tumor microenvironment. In this study, we assessed how hemin, a pharmacologic inducer of heme oxygenase-1 (HO-1), has an impact on prostate cancer development in an in vivo conditioning model.Experimental Design: The stroma of C57BL/6 mice was conditioned by subcutaneous administration of hemin prior to TRAMP-C1 tumor challenge. Complementary in vitro and in vivo assays were performed to evaluate hemin effect on both angiogenesis and the immune response. To gain clinical insight, we used prostate cancer patient-derived samples in our studies to assess the expression of HO-1 and other relevant genes.Results: Conditioning resulted in increased tumor latency and decreased initial growth rate. Histologic analysis of tumors grown in conditioned mice revealed impaired vascularization. Hemin-treated human umbilical vein endothelial cells (HUVEC) exhibited decreased tubulogenesis in vitro only in the presence of TRAMP-C1–conditioned media. Subcutaneous hemin conditioning hindered tumor-associated neovascularization in an in vivo Matrigel plug assay. In addition, hemin boosted CD8+ T-cell proliferation and degranulation in vitro and antigen-specific cytotoxicity in vivo. A significant systemic increase in CD8+ T-cell frequency was observed in preconditioned tumor-bearing mice. Tumors from hemin-conditioned mice showed reduced expression of galectin-1 (Gal-1), key modulator of tumor angiogenesis and immunity, evidencing persistent remodeling of the microenvironment. We also found a subset of prostate cancer patient-derived xenografts and prostate cancer patient samples with mild HO-1 and low Gal-1 expression levels.Conclusions: These results highlight a novel function of a human-used drug as a means of boosting the antitumor response. Clin Cancer Res; 23(17); 5135–48. ©2017 AACR.
- Published
- 2023
15. Figure S1 from In Vivo Hemin Conditioning Targets the Vascular and Immunologic Compartments and Restrains Prostate Tumor Development
- Author
-
Elba S. Vazquez, Diego J. Laderach, Daniel Compagno, Gabriel A. Rabinovich, Nora Navone, Asif Ahmed, Paula M. Berguer, Emiliano G. Ortiz, Roberto P. Meiss, Geraldine Gueron, Lucas D. Gentilini, and Felipe M. Jaworski
- Abstract
In vitro tubulogenesis assay. A, in vitro tube formation using control or hemin-treated HUVEC in the absence or presence of T-C1 conditioned media. Complete growth media served as a positive control. Figure depicts one representative out of three independent experiments. B, total tube length was quantified. *** indicates P
- Published
- 2023
16. Table S1 from In Vivo Hemin Conditioning Targets the Vascular and Immunologic Compartments and Restrains Prostate Tumor Development
- Author
-
Elba S. Vazquez, Diego J. Laderach, Daniel Compagno, Gabriel A. Rabinovich, Nora Navone, Asif Ahmed, Paula M. Berguer, Emiliano G. Ortiz, Roberto P. Meiss, Geraldine Gueron, Lucas D. Gentilini, and Felipe M. Jaworski
- Abstract
List of the primers used in qPCR analyses carried out in this study.
- Published
- 2023
17. Table S4 from Targeting the MYCN–PARP–DNA Damage Response Pathway in Neuroendocrine Prostate Cancer
- Author
-
Timothy C. Thompson, Patricia Troncoso, Nora Navone, Arul M. Chinnaiyan, Ana M. Aparicio, Christopher J. Logothetis, Paul G. Corn, Jeri Kim, Xuhong Cao, Abul Kalam Azad, Sanghee Park, Guang Yang, Jianxiang Wang, Yang Luan, Dimitrios Korentzelos, Spyridon P. Basourakos, Bradley M. Broom, Likun Li, Wenhui Wu, Bo Liu, and Wei Zhang
- Abstract
Differential expression results and pathway distribution for common significant DDR-M genes in Beltran's and PDX RNAseq datasets
- Published
- 2023
18. Figure S2 from In Vivo Hemin Conditioning Targets the Vascular and Immunologic Compartments and Restrains Prostate Tumor Development
- Author
-
Elba S. Vazquez, Diego J. Laderach, Daniel Compagno, Gabriel A. Rabinovich, Nora Navone, Asif Ahmed, Paula M. Berguer, Emiliano G. Ortiz, Roberto P. Meiss, Geraldine Gueron, Lucas D. Gentilini, and Felipe M. Jaworski
- Abstract
Expression of CD146 mRNA levels in control and hemin-treated BAEC (50 μM, 8 h), as determined by RT-qPCR. Bovine GAPDH was used as an internal reference gene. * indicates P
- Published
- 2023
19. Supplementary Tables from Radium-223 Treatment Increases Immune Checkpoint Expression in Extracellular Vesicles from the Metastatic Prostate Cancer Bone Microenvironment
- Author
-
Theocharis Panaretakis, Christopher J. Logothetis, Sue-Hwa Lin, Sumit K. Subudhi, Eleni Efstathiou, Gary E. Gallick, Nora Navone, Patricia Troncoso, Marites P. Melancon, Elmer Santos, Guoyu Yu, Song-Chang Lin, Yu-Chen Lee, Leah Guerra, Nila Parikh, Anh Hoang, Namrata Madan, Jian H. Song, Emanuela Gentile, Paul Corn, and Ioulia Vardaki
- Abstract
Supp Tables 1-3
- Published
- 2023
20. Table S2 from In Vivo Hemin Conditioning Targets the Vascular and Immunologic Compartments and Restrains Prostate Tumor Development
- Author
-
Elba S. Vazquez, Diego J. Laderach, Daniel Compagno, Gabriel A. Rabinovich, Nora Navone, Asif Ahmed, Paula M. Berguer, Emiliano G. Ortiz, Roberto P. Meiss, Geraldine Gueron, Lucas D. Gentilini, and Felipe M. Jaworski
- Abstract
Expression microarray studies available in the Oncomine database that met our eligibility criteria.
- Published
- 2023
21. Figure S1 from Targeting the MYCN–PARP–DNA Damage Response Pathway in Neuroendocrine Prostate Cancer
- Author
-
Timothy C. Thompson, Patricia Troncoso, Nora Navone, Arul M. Chinnaiyan, Ana M. Aparicio, Christopher J. Logothetis, Paul G. Corn, Jeri Kim, Xuhong Cao, Abul Kalam Azad, Sanghee Park, Guang Yang, Jianxiang Wang, Yang Luan, Dimitrios Korentzelos, Spyridon P. Basourakos, Bradley M. Broom, Likun Li, Wenhui Wu, Bo Liu, and Wei Zhang
- Abstract
Expression of MYCN-PARP-DDR pathway genes in MYCN neutral and amplified neuroblastoma tumor samples.
- Published
- 2023
22. Data from PCAT-1, a Long Noncoding RNA, Regulates BRCA2 and Controls Homologous Recombination in Cancer
- Author
-
Felix Y. Feng, Arul M. Chinnaiyan, Theodore S. Lawrence, Francesca Demichelis, Mark A. Rubin, Naoki Kitabayashi, Karen E. Knudsen, Christine E. Canman, Ashutosh K. Tewari, Louis L. Pisters, John C. Araujo, Christopher J. Logothetis, Nora Navone, Kari Wilder-Romans, Rohit Malik, Anirban Sahu, Sumin Han, Teng Ma, Qi Cao, Matthew K. Iyer, Wei Chen, and John R. Prensner
- Abstract
Impairment of double-stranded DNA break (DSB) repair is essential to many cancers. However, although mutations in DSB repair proteins are common in hereditary cancers, mechanisms of impaired DSB repair in sporadic cancers remain incompletely understood. Here, we describe the first role for a long noncoding RNA (lncRNA) in DSB repair in prostate cancer. We identify PCAT-1, a prostate cancer outlier lncRNA, which regulates cell response to genotoxic stress. PCAT-1 expression produces a functional deficiency in homologous recombination through its repression of the BRCA2 tumor suppressor, which, in turn, imparts a high sensitivity to small-molecule inhibitors of PARP1. These effects reflected a posttranscriptional repression of the BRCA2 3′UTR by PCAT-1. Our observations thus offer a novel mechanism of “BRCAness” in sporadic cancers. Cancer Res; 74(6); 1651–60. ©2014 AACR.
- Published
- 2023
23. Characterization of prostate cancer adrenal metastases: dependence upon androgen receptor signaling and steroid hormones
- Author
-
Minas J. Sakellakis, Andrew W. Hahn, Sumankalai Ramachandran, Miao Zhang, Anh Hoang, Jian H. Song, Jingjing Liu, Feng Wang, Hirak S. Basu, Peter Sheperd, Xuemei Wang, Daniel E. Frigo, Sue-Hwa Lin, Theocharis Panaretakis, Jianhua Zhang, Nora Navone, Patricia Troncoso, Christopher J. Logothetis, and Mark A. Titus
- Subjects
Cancer Research ,Oncology ,Urology - Abstract
Prostate cancer (PCa) typically spreads to the bone, and this distribution is attributed to the central role of the microenvironment in progression. However, metastasis to the adrenal glands, while not as common, does occur. The biology that accounts for adrenal metastases may be attributed to the unique local steroid metabolome and co-clinical characterization may elucidate the role steroid biosynthesis plays in PCa progression.Three patients with metastatic PCa who had archived tumor tissue from an adrenalectomy were retrospectively identified, and one adrenal metastasis was developed into a xenograft (MDA-PCa-250). The adrenal metastases were characterized by performing somatic DNA whole exome sequencing (WES), RNA-Seq, immunohistochemistry (IHC), and steroid metabolite quantitation. The influence of steroid metabolites on adrenal metastasis cells and tumor growth was tested in vitro and in vivo.Clinically, adrenalectomy was performed during castration-resistant oligometastatic disease, and two men experienced resensitization to leuprolide. Somatic DNA WES revealed heterogeneous alterations in tumor suppressor and DNA damage repair pathway genes. Adrenal metastases had active androgen receptor (AR) signaling by IHC, and RNA-Seq supported a potential role for adrenal androgen precursor metabolism in activating the AR. Steroid quantitation suggested the adrenal androgen precursors were converted into testosterone in these metastases, and stable isotope tracing of an organoid from MDA-PCa-250 confirmed the capability of adrenal metastases to biosynthesize testosterone from adrenal precursors. In vitro testing of a cell line derived from MDA-PCa-250 showed that testosterone and cortisol stimulated tumor cell growth. In vivo experiments demonstrated that MDA-PCa-250 grew in intact mice with circulating testosterone, but not in castrated mice.PCa adrenal metastases depend upon AR signaling driven by androgen precursors, androstenedione and dehydroepiandrosterone, available in the microenvironment, despite the presence of heterogeneous somatic DNA alterations. Moreover, MDA-PCa-250 provides a preclinical model that can recapitulate the unique androgen-dependence of adrenal metastases.This study does not report the clinical results of a clinical trial, but it does use samples from a completed clinical trial that is registered with clinicaltrials.gov (NCT01254864).
- Published
- 2022
24. Pin-pointing the key hubs in the IFN-γ pathway responding to SARS-CoV-2 infection
- Author
-
Ayelen Toro, Sofia Lage-Vickers, Juan Bizzotto, Felipe Vilicich, Agustina Sabater, Gaston Pascual, Sabrina Ledesma-Bazan, Pablo Sanchis, Maria S. Ruiz, Ana P. Arevalo, Jorge L. Porfido, Rocio Seniuk, Estefania Labanca, Nicolas Anselmino, Nora Navone, Daniel F. Alonso, Elba Vazquez, Martina Crispo, Javier Cotignola, and Geraldine Gueron
- Abstract
Interferon gamma may be a potential adjuvant immunotherapy for COVID-19 patients. In this work, we assessed gene expression profiles associated with the IFN-γ pathway in response to SARS-CoV-2 infection. Employing a case-control study from SARS-CoV-2 positive and negative patients, we identified IFN-γ-associated pathways to be enriched in positive patients. Bioinformatics analyses showed upregulation of MAP2K6, CBL, RUNX3, STAT1 and JAK2 in COVID-19 positive vs. negative patients. A positive correlation was observed between STAT1/JAK2, which varied alongside the patient’s viral load. Expression of MX1, MX2, ISG15 and OAS1 (4 well-known IFN-stimulated genes (ISGs)) displayed upregulation in COVID-19 positive vs. negative patients. Integrative analyses showcased higher levels of ISGs which were associated with increased viral load and STAT1/JAK2 expression. Confirmation of ISGs up-regulation was performed in vitro using the A549 lung cell line treated with Poly(I:C), a synthetic analog of viral double-stranded RNA; and in different pulmonary human cell lines and ferret tracheal biopsies infected with SARS-CoV-2. A pre-clinical murine model of coronavirus infection confirmed findings displaying increased ISGs in the liver and lungs from infected mice. Altogether, these results demonstrate the role of IFN-γ and ISGs in response to SARS-CoV-2 infection, highlighting alternative druggable targets that can boost the host response.
- Published
- 2022
25. KDM4A promotes the progression of neuroendocrine prostate cancer
- Author
-
Celia Sze Ling Mak, Ming Zhu, Xin Liang, Feng Wang, Fei Yuan, Anh G Hoang, Xingzhi Song, Peter Shepherd, Derek Liang, Jessica Suh, Bijeta Pradhan, Jiwon Park, Miao Zhang, Eric Metzger, Roland Schüle, Abhinav K. Jain, Ellen Karasik, Barbara A. Foster, Min Gyu Lee, Paul Corn, Christopher J. Logothetis, Ana Aparicio, Nora Navone, Patricia Troncoso, Zhi Tan, Jianhua Zhang, Sue-Hwa Lin, and Guocan Wang
- Abstract
Neuroendocrine prostate cancer (NEPC) represents one of the most lethal forms of prostate cancer (PCa) and lacks life-prolonging treatment. The incidence of NEPC is increased due to the widespread use of AR pathway inhibitors (ARPIs) in the treatment of non-metastatic CRPC and hormone-sensitive metastatic tumors. Here, we identified histone lysine demethylase KDM4A as a key player in NEPC progression and an effective therapeutic target. We found that KDM4A mRNA and protein are overexpressed in human and mouse NEPC compared to prostate adenocarcinoma. Knockdown (KD) or knockout (KO) ofKDM4Ain NEPC cell lines suppressed cancer cell growthin vitroandin vivo. Mechanistically, we found that KDM4A promotes NEPC progression, in part, through direct transcriptional regulation ofMYC. We showed thatMYCis hyper-activated in human and mouse NEPC.KDM4AKD led to suppression of MYC signaling.MYCKD or inhibition profoundly suppressed NEPC cell proliferation. Furthermore, a potent pan-KDM4 inhibitor QC6352 significantly reduced NEPC cell growthin vitroandin vivo. Taken together, we demonstrated that KDM4A is an important regulator of NEPC progression and targeting KDM4A may potentially be an effective therapeutic strategy for NEPC.SignificanceNeuroendocrine prostate cancer (NEPC) is a highly aggressive prostate cancer subtype that is resistant to potent androgen receptor pathway inhibitors (ARPIs) and currently lacks effective therapeutic options. Histone lysine demethylase KDM4A is an important epigenetic regulator of gene expression in development and cancer. In this study, we show that KDM4A is highly expressed in NEPC and is required for NEPC proliferation, anchorage-independent growth, andin vivogrowth, which is in part mediated through the regulation of MYC expression. Importantly, we demonstrate that inhibition of KDM4A significantly impairs NEPC growth in preclinical models. Thus, our findings provide valuable insights into the molecular mechanisms underlying NEPC progression and offer a rationale for clinical trials with KDM4 inhibitor in NEPC patients.
- Published
- 2022
26. Abstract 142: Candidate measures of lineage plasticity in aggressive phenotypes of prostate cancer
- Author
-
Souzana Logotheti, Haswanth Vundavilli, Eugenia Papadaki, Yuehui Zhao, Marcos Roberto Estecio, Rama Soundararajan, Peter Shepherd, Jiabin Dong, Anh Hoang, Shuai Guo, Nora Navone, Vasiliki Tzelepi, Christopher Logothetis, Yue Lu, Aristidis Vrahatis, Wenyi Wang, and Ana Aparicio
- Subjects
Cancer Research ,Oncology - Abstract
Lineage plasticity in cancer reflects the property of neoplastic cells to adapt to external pressures by switching between different developmental pathways. While some prostate cancers appear “constrained” to a luminal epithelial lineage despite progression on androgen signaling inhibitors, others exhibit a “lineage-promiscuous” phenotype under the pressures of these therapies. However, a clinically applicable marker that quantifies lineage plasticity has yet to be developed. To address this knowledge gap, we applied single-cell genomics and bulk epigenetic profiling to two patient derived xenografts (PDXs) derived before and after chemotherapy from the castration-resistant prostate tumor of a 42-year-old with a PSA 486, Gleason 5+4, cT4N1M1b PCa, who succumbed to his disease within 21 months of diagnosis. We reasoned that the two PDXs, MDA PCa 177-B (AR negative, basal transcriptional profile) and MDA PCa 189-1 (AR positive, luminal transcriptional profile), should share the property of lineage plasticity and serve to define candidate measurable signatures of this property. Targeted and single-cell DNA sequencing showed that the two models share 66.8% of their annotated mutations, 71.5% of copy number variations, and a common ancestor in their phylogeny, confirming their common clonal origin. Single-cell RNA sequencing revealed the presence of a cluster of cells shared by both PDXs that contained a highly mixed program, similar to a “high-plasticity cell state” (HPCS) previously implicated in the emergence and maintenance of cellular heterogeneity in genetically engineered mouse models of lung cancer (Marjanovic et al PMID: 32707077). H3K27ac, H3K27me3 and H3K4me3 chromatin immunoprecipitation followed by sequencing and reduced representation bisulfite sequencing performed in triplicates revealed chromatin profiles shared by both PDX models, and being absent from normal prostate specimens in publicly available datasets. In conclusion, our data supports the hypothesis that the property of lineage plasticity can be reflected in a biomarker signature and used as a measurable metric. Ongoing are evaluation of the candidate signatures in publicly available datasets representative of the spectrum of the disease in patient samples, and assessment of the effects of systemic therapies (including epigenetic modulators) on the property of lineage plasticity in clinical and co-clinical samples. Citation Format: Souzana Logotheti, Haswanth Vundavilli, Eugenia Papadaki, Yuehui Zhao, Marcos Roberto Estecio, Rama Soundararajan, Peter Shepherd, Jiabin Dong, Anh Hoang, Shuai Guo, Nora Navone, Vasiliki Tzelepi, Christopher Logothetis, Yue Lu, Aristidis Vrahatis, Wenyi Wang, Ana Aparicio. Candidate measures of lineage plasticity in aggressive phenotypes of prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 142.
- Published
- 2023
27. Abstract 4760: KDM4A promotes NEPC progression through regulation of MYC expression
- Author
-
Celia Sze Ling Mak, Ming Zhu, Xin Liang, Feng Wang, Anh G. Hoang, Xinzhi Song, Peter Shepherd, Derek Liang, Jessica Suh, Jiwon Park, Miao Zhang, Eric Metzger, Roland Schule, Abhinav K. Jain, Ellen Karasik, Barbara A. Foster, Min Gyu Lee, Paul Corn, Christopher J. Logothetis, Ana Aparicio, Nora Navone, Patricia Troncoso, Jianhua Zhang, Sue-Hwa Lin, and Guocan Wang
- Subjects
Cancer Research ,Oncology - Abstract
Despite advancements in treatment, prostate cancer (PCa) remains the second leading cause of death among men. Neuroendocrine prostate cancer (NEPC) represents one of the most lethal forms of PCa and lacks life-prolonging treatment. Here we identified histone lysine demethylase KDM4A as a driver in NEPC progression and an effective therapeutic target. KDM4A mRNA and protein are overexpressed in human and mouse NEPC compared to adenocarcinoma. Knockdown or knockout of KDM4A in NEPC cell lines suppressed cancer cell growth in vitro and in vivo. Importantly, the inactivation of Kdm4a in a genetically engineered mouse model of prostate cancer reduces tumor burden, reduces the incidence of NEPC, and prolongs overall survival. Mechanistically, KDM4A directly regulates the transcription of MYC, which is hyper-activated in human and mouse NEPC. Furthermore, a potent pan-KDM4 inhibitor QC6352 significantly reduces NEPC cell growth in vitro and in vivo. Taken together, we demonstrate that KDM4A promotes NEPC progression through regulation of MYC expression and targeting KDM4A can be an effective therapeutic strategy for NEPC. Citation Format: Celia Sze Ling Mak, Ming Zhu, Xin Liang, Feng Wang, Anh G. Hoang, Xinzhi Song, Peter Shepherd, Derek Liang, Jessica Suh, Jiwon Park, Miao Zhang, Eric Metzger, Roland Schule, Abhinav K. Jain, Ellen Karasik, Barbara A. Foster, Min Gyu Lee, Paul Corn, Christopher J. Logothetis, Ana Aparicio, Nora Navone, Patricia Troncoso, Jianhua Zhang, Sue-Hwa Lin, Guocan Wang. KDM4A promotes NEPC progression through regulation of MYC expression. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4760.
- Published
- 2023
28. Targeting histone lysine demethylase KDM4A in Aggressive Variant Prostate Cancer
- Author
-
Celia Sze Ling Mak, Ming Zhu, Xin Liang, Feng Wang, Anh G. Hoang, Xinzhi Song, Peter Shepherd, Derek Liang, Jessica Suh, Jiwon Park, Miao Zhang, Eric Metzger, Roland Shule, Abhinav K. Jain, Ellen Karasik, Barbara A. Foster, Min Gyu Lee, Paul Corn, Christopher J. Logothetis, Ana Aparicio, Nora Navone, Patricia Troncoso, Jianhua Zhang, Sue-Hwa Lin, and Guocan Wang
- Published
- 2022
29. Exploiting Interdata Relationships in Prostate Cancer Proteomes: Clinical Significance of HO-1 Interactors
- Author
-
Sofia Lage-Vickers, Pablo Sanchis, Juan Bizzotto, Ayelen Toro, Agustina Sabater, Rosario Lavignolle, Nicolas Anselmino, Estefania Labanca, Alejandra Paez, Nora Navone, Maria P. Valacco, Javier Cotignola, Elba Vazquez, and Geraldine Gueron
- Subjects
Physiology ,Clinical Biochemistry ,YWHAZ ,HMOX1 ,prostate cancer ,proteomics ,transcriptomics ,protein interactions ,Cell Biology ,Molecular Biology ,Biochemistry - Abstract
Prostate cancer (PCa) cells display abnormal expression of proteins resulting in an augmented capacity to resist chemotherapy and colonize distant organs. We have previously shown the anti-tumoral role of heme oxygenase 1 (HO-1) in this disease. In this work, we undertook a mass spectrometry-based proteomics study to identify HO-1 molecular interactors that might collaborate with its modulatory function in PCa. Among the HO-1 interactors, we identified proteins with nuclear localization. Correlation analyses, using the PCa GSE70770 dataset, showed a significant and positive correlation between HMOX1 and 6 of those genes. Alternatively, HMOX1 and YWHAZ showed a negative correlation. Univariable analyses evidenced that high expression of HNRNPA2B1, HSPB1, NPM1, DDB1, HMGA1, ZC3HAV1, and HMOX1 was associated with increased relapse-free survival (RFS) in PCa patients. Further, PCa patients with high HSPB1/HMOX1, DDB1/HMOX1, and YWHAZ/HMOX1 showed a worse RFS compared with patients with lower ratios. Moreover, a decrease in RFS for patients with higher scores of this signature was observed using a prognostic risk score model. However, the only factor significantly associated with a higher risk of relapse was high YWHAZ. Multivariable analyses confirmed HSPB1, DDB1, and YWHAZ independence from PCa clinic-pathological parameters. In parallel, co-immunoprecipitation analysis in PCa cells ascertained HO-1/14-3-3ζ/δ (protein encoded by YWHAZ) interaction. Herein, we describe a novel protein interaction between HO-1 and 14-3-3ζ/δ in PCa and highlight these factors as potential therapeutic targets.
- Published
- 2021
30. Abstract 2374: Early metabolic rewiring of prostate cancer cells triggered by bone progenitors defines survival of metastatic prostate cancer
- Author
-
Pablo Sanchis, Nicolas Anselmino, Rosario Lavignolle, Agustina Sabater, Estefania Labanca, Juan Bizzotto, Sofia Lage-Vickers, Gaston Pascual, Rocio Seniuk, Ayelen Toro, Nora Navone, Javier Cotignla, Elba Vazquez, and Geraldine Gueron
- Subjects
Cancer Research ,Oncology - Abstract
Bone hosts the 88% of Prostate Cancer (PCa) metastases and no curative therapy is currently available for this stage. Arrival of PCa cells to the bone homing organ is accompanied by a metabolic adaptation, which may be mediated by bone secreted factors. Herein, we sought to identify key metabolic genes fueling PCa bone metastasis and soluble factors secreted by bone cells leading to the metabolic rewiring of tumoral cells. By an indirect transwell co-culture system of PCa (PC3) and bone progenitor cells (MC3T3, pre-osteoblasts; or Raw264.7, pre-osteoclasts) we analyzed the transcriptome (RNA-seq) of PC3 cells modulated by soluble factors released from bone precursors. GSEA showed a strong activation of lipid metabolism, including PPAR and PI3K-Akt pathways, fat absorption and digestion. We then performed a Principal Component Analysis using transcriptomic data from human PCa and bone metastasis samples (GSE74685), showcasing that those metabolic genes that appeared significantly dysregulated in the co-culture model could accurately cluster samples by their tissue of origin in two defined groups: primary PCa and bone metastasis. This result was confirmed by an unsupervised clustering analysis, highlighting that the transcriptional metabolic profile triggered in the in vitro model has a clinical correlate in human bone metastasis samples. Interestingly, when performing a survival analysis for those genes in the SU2C-PCF dataset, we observed that 4 lipid-associated genes, PPARA, VDR, SLC16A1 and GPX1, correlated with a shorter overall survival time, and appeared as independent risk-predictors of death (HR: 4.96, 2.85, 3.93 and 3.67, respectively; P Citation Format: Pablo Sanchis, Nicolas Anselmino, Rosario Lavignolle, Agustina Sabater, Estefania Labanca, Juan Bizzotto, Sofia Lage-Vickers, Gaston Pascual, Rocio Seniuk, Ayelen Toro, Nora Navone, Javier Cotignla, Elba Vazquez, Geraldine Gueron. Early metabolic rewiring of prostate cancer cells triggered by bone progenitors defines survival of metastatic prostate 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 2374.
- Published
- 2022
31. Abstract 284: A 3D multicellular in vitro prostate cancer model featuring racially/ethnically diverse PDXs
- Author
-
Divya Iyer, Andrei Bonteanu, Peter Shepherd, Rick Kittles, Nora Navone, Daniel Harrington, and Kristin Bircsak
- Subjects
Cancer Research ,Oncology - Abstract
Current in vitro prostate cancer (PCa) research tools do not incorporate the complexities of the tumor microenvironment including perfusion, multiple cell types, extracellular matrix (ECM) and 3D-orientation. Additionally, these models lack racial/ethnic (R/E) diversity, failing to address the cancer health disparity (CHD) observed in Black men with PCa who experience double the incidence and mortality rates as compared to White men. Population-based in vitro PCa models that support crosstalk between different cell types will improve our understanding of the underlying mechanisms contributing to this CHD and improve the prediction of drug response in specific populations. Here, we used the high-throughput, perfusion-based microfluidic platform called the MIMETAS OrganoPlate® to culture up to 96 individual, multicellular PCa-on-a-chip cultures in parallel. PCa PDX cells of various R/E were embedded together with E2Crimson-labeled stromal fibroblasts in a migration-permissive hyaluronic acid hydrogel in the gel compartment, alongside an endothelium-lined perfusion channel containing a targeted immune cell population. Using high content imaging, we confirmed the stability and viability of the cultures over 7 days. The cultures maintained a 3D structure with PCa cells and fibroblasts evenly dispersed throughout the height of the gel compartment and a blood vessel-like structure within the perfusion channel. Closer evaluation of the gel compartment revealed a close association between PCa cells and stromal fibroblasts in a core-shell structure with PCa clusters surrounded by fibroblasts. Further, all cells maintained expected expression of phenotypic markers (PCa: epCAM, PSMA; fibroblast: vimentin, endothelium: CD31). For the immune component, medium and cell tracker dye conditions were optimized to support immune cell viability. Over 72 hours, immune cells were monitored, revealing migration of PBMCs through the gel compartment. Finally, the value of the complex, population-based multicellular in vitro model of PCa will be evaluated in a drug screen comparing the system with simple, PCa PDX monocultures. This R/E diverse 3D prostate tumor model will enable the full incorporation of all cell types and ECM into a single model which will better recapitulate population specific PCa and drug response. Citation Format: Divya Iyer, Andrei Bonteanu, Peter Shepherd, Rick Kittles, Nora Navone, Daniel Harrington, Kristin Bircsak. A 3D multicellular in vitro prostate cancer model featuring racially/ethnically diverse PDXs [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 284.
- Published
- 2022
32. Protein Kinase A (PKA) as a Master Regulator of the Early Metabolic Reprogramming in Bone Metastatic Prostate Cancer Cells
- Author
-
Pablo Sanchis, Nicolas Anselmino, Rosario Lavignolle, Agustina Sabater, Estefania Labanca, Juan Bizzotto, Sofia Lage-Vickers, Gaston Pascual, Rocio Seniuk, Ayelen Toro, Nora Navone, Javier Cotignola, Elba Vazquez, and Geraldine Gueron
- Subjects
Cancer Research ,Oncology - Abstract
PURPOSE The arrival of metastatic prostate cancer (PCa) tumor cells to the bone niche requires a metabolic adaptation. We sought to identify metabolic dysregulations fueling PCa metastasis, modulated by bone secreted factors. METHODS By an indirect co-culture system of PCa (PC3) and bone progenitors (MC3T3, pre-osteoblasts, or Raw 264.7, pre-osteoclasts) we assessed the transcriptome of PC3 cells modulated by soluble factors released from bone precursors. We validated the transcriptional profile of metabolic genes in open-access transcriptomic datasets. We performed an Ingenuity Pathway Analysis (IPA) to delineate the regulators of these metabolic genes. Bone secretome was profiled on the conditioned media (CM) by ESI-MS/MS. RESULTS PC3 cells co-cultured with bone progenitors displayed an activation of lipidic categories, including PPAR-signaling and fat absorption/digestion. Principal Component and Unsupervised Clustering analyses using transcriptomic data from human PCa and bone metastatic samples (GSE74685) showed that the metabolic genes deregulated in PC3 accurately clustered samples in primary tumor or bone metastasis. Moreover, four lipid-associated genes, PPARA, VDR, SLC16A1 and GPX1, were associated with a shorter survival time (SU2C-PCF dataset), and were independent risk-predictors of death ( P < .05). An IPA revealed that these genes are regulated by the Protein Kinase A (PKA). Accordingly, PC3 cells treated with the CM of the co-culture presented a decreased ATP content compared to the treatment with the CM of PC3 grown alone, which was restored upon PKA inhibition. Finally, the secretome analysis revealed soluble factors secreted by bone progenitors (Col1a1, Fn1) which could regulate PKA activity. CONCLUSION We identified a novel lipid-associated gene signature important for metastatic PCa, triggered by the dialogue with bone cells. Moreover, PKA could regulate this signature in response to bone-secreted factors reprogramming the metabolic phenotype of metastatic cells, emerging as a potential druggable target for this disease.
- Published
- 2022
33. Transcriptional Inactivation of TP53 and the BMP Pathway Mediates Therapy-induced Dedifferentiation and Metastasis in Prostate Cancer
- Author
-
Hyunho Han, Yan Wang, Josue Curto, Sreeharsha Gurrapu, Sara Laudato, Alekya Rumandla, Goutam Chakraborty, Xiaobo Wang, Hong Chen, Yan Jiang, Dhiraj Kumar, Emily Caggiano, Boyu Zhang, Yan Ji, Sankar N. Maity, Min Hu, Shanshan Bai, Ana Aparicio, Eleni Efstathiou, Christopher J. Logothetis, Nicholas Navin, Nora Navone, Yu Chen, and Filippo G. Giancotti
- Abstract
SummaryUnsupervised clustering and deconvolution analysis identifies a novel subtype of M-CRPC endowed with hybrid epithelial/mesenchymal (E/M) and luminal progenitor-like traits (Mesenchymal and Stem-like PC, MSPC). Analysis of patient datasets and mechanistic studies indicate that MSPC arises as a consequence of therapy-induced lineage plasticity. AR blockade instigates two separate and complementary processes: 1) transcriptional silencing of TP53 and hence acquisition of hybrid E/M and stem-like traits; and 2) inhibition of the BMP signaling, which promotes resistance to the pro-apoptotic and anti-proliferative effects of AR inhibition. The drug-tolerant prostate cancer cells generated through reprogramming are rescued by neuregulin and generate metastases in mice. Combined inhibition of HER2/3 and AR or mTORC1 exhibit efficacy in preclinical models of mixed ARPC/MSPC or MSPC, respectively. These results identify a novel subtype of M-CRPC, trace its origin to therapy-induced lineage plasticity, and reveal its dependency on HER2/3 signaling.
- Published
- 2021
34. Optimizing the diagnosis and management of ductal prostate cancer
- Author
-
Weranja, Ranasinghe, Daniel D, Shapiro, Miao, Zhang, Tharakeswara, Bathala, Nora, Navone, Timothy C, Thompson, Bradley, Broom, Ana, Aparicio, Shi-Ming, Tu, Chad, Tang, John W, Davis, Louis, Pisters, and Brian F, Chapin
- Subjects
Carcinoma, Ductal ,Male ,Humans ,Prostatic Neoplasms - Abstract
Ductal adenocarcinoma (DAC) is the most common variant histological subtype of prostate carcinoma and has an aggressive clinical course. DAC is usually characterized and treated as high-risk prostatic acinar adenocarcinoma (PAC). However, DAC has a different biology to that of acinar disease, which often poses a challenge for both diagnosis and management. DAC can be difficult to identify using conventional diagnostic modalities such as serum PSA levels and multiparametric MRI, and the optimal management for localized DAC is unknown owing to the rarity of the disease. Following definitive therapy for localized disease with radical prostatectomy or radiotherapy, the majority of DACs recur with visceral metastases at low PSA levels. Various systemic therapies that have been shown to be effective in high-risk PAC have limited use in treating DAC. Although current understanding of the biology of DAC is limited, genomic analyses have provided insights into the pathology behind its aggressive behaviour and potential future therapeutic targets.
- Published
- 2021
35. Enhanced Viability for Ex vivo 3D Hydrogel Cultures of Patient-Derived Xenografts in a Perfused Microfluidic Platform
- Author
-
Daniel A. Harrington, Nora Navone, Anthony Saleh, Pamela E. Constantinou, Mary C. Farach-Carson, Karla Queiroz, Peter Shepherd, Kristin M. Bircsak, and Lindsey K. Sablatura
- Subjects
General Immunology and Microbiology ,General Chemical Engineering ,General Neuroscience ,General Biochemistry, Genetics and Molecular Biology - Published
- 2020
36. Bone Progenitors Pull the Strings on the Early Metabolic Rewiring Occurring in Prostate Cancer Cells
- Author
-
Pablo Sanchis, Nicolas Anselmino, Sofia Lage-Vickers, Agustina Sabater, Rosario Lavignolle, Estefania Labanca, Peter D. A. Shepherd, Juan Bizzotto, Ayelen Toro, Antonina Mitrofanova, Maria Pia Valacco, Nora Navone, Elba Vazquez, Javier Cotignola, and Geraldine Gueron
- Subjects
Cancer Research ,Oncology ,prostate cancer ,bone metastasis ,metabolism ,gene signature ,lipid metabolism ,PKA ,urologic and male genital diseases - Abstract
Metastatic prostate cancer (PCa) cells soiling in the bone require a metabolic adaptation. Here, we identified the metabolic genes fueling the seeding of PCa in the bone niche. Using a transwell co-culture system of PCa (PC3) and bone progenitor cells (MC3T3 or Raw264.7), we assessed the transcriptome of PC3 cells modulated by soluble factors released from bone precursors. In a Principal Component Analysis using transcriptomic data from human PCa samples (GSE74685), the altered metabolic genes found in vitro were able to stratify PCa patients in two defined groups: primary PCa and bone metastasis, confirmed by an unsupervised clustering analysis. Thus, the early transcriptional metabolic profile triggered in the in vitro model has a clinical correlate in human bone metastatic samples. Further, the expression levels of five metabolic genes (VDR, PPARA, SLC16A1, GPX1 and PAPSS2) were independent risk-predictors of death in the SU2C-PCF dataset and a risk score model built using this lipid-associated signature was able to discriminate a subgroup of bone metastatic PCa patients with a 23-fold higher risk of death. This signature was validated in a PDX pre-clinical model when comparing MDA-PCa-183 growing intrafemorally vs. subcutaneously, and appears to be under the regulatory control of the Protein Kinase A (PKA) signaling pathway. Secretome analyses of conditioned media showcased fibronectin and type-1 collagen as critical bone-secreted factors that could regulate tumoral PKA. Overall, we identified a novel lipid gene signature, driving PCa aggressive metastatic disease pointing to PKA as a potential hub to halt progression.
- Published
- 2022
37. Oncogenic and osteolytic functions of histone demethylase NO66 in castration-resistant prostate cancer
- Author
-
Krishna M, Sinha, Rozita, Bagheri-Yarmand, Sharmistha, Lahiri, Yue, Lu, Miao, Zhang, Sarah, Amra, Yasmeen, Rizvi, Xinhai, Wan, Nora, Navone, Bulent, Ozpolat, Christopher, Logothetis, Robert F, Gagel, and Johnny, Huard
- Subjects
Histone Demethylases ,Male ,Bone Neoplasms ,Mice, SCID ,Osteolysis ,Dioxygenases ,Epigenesis, Genetic ,Gene Expression Regulation, Neoplastic ,Histones ,Mice ,Prostatic Neoplasms, Castration-Resistant ,Cell Transformation, Neoplastic ,HEK293 Cells ,Cell Line, Tumor ,PC-3 Cells ,NIH 3T3 Cells ,Animals ,Humans - Abstract
Epigenetic changes that cause dysregulated gene expression during progression of androgen-independent prostate cancer (PCa) and metastatic skeletal lesions remain elusive. Here, we explored the role of histone demethylase NO66 in the pathogenesis of PCa and bone metastasis-related skeletal lesions. Tissue and cDNA microarrays of PCa were analyzed for NO66 mRNA and protein levels. We examined the effects of gain and loss of NO66 function on cell viability, colony formation, migration, invasion, and tumor-induced skeletal lesions in femoral bone. RNAseq and ChIPseq were performed to elucidate NO66-target genes in PCa. We report that NO66 levels were upregulated in advanced primary prostate tumors compared to normal tissue or tumors with low Gleason scores. Forced expression of NO66 promoted cell survival and invasion of PCa cells; whereas, knockdown of NO66 resulted in decreased cell survival and increased sensitivity to docetaxel. NO66-overexpressing PC3 cells implanted into the femoral bone of male SCID mice caused massive bone loss and stimulation of mouse osteoclast-promoting genes, including Dickkopf1, Cathepsin K, Nf-kβ,; and Calcr, suggesting a role for NO66 in tumor growth in bone and osteoclast activity. Combined RNAseq and ChIP-seq revealed that NO66 activates the survival gene MCL1, the invasion-associated genes IGFBP5 and MMP3, the pro-oncogenic genes CTNNB1 and CCND1, and the epigenetic modifier gene KMT2A in androgen-independent PCa. Our findings uncover the role of NO66 as a key oncogenic driver in PCa, causing osteolytic lesions through upstream epigenetic regulation of key genes for survival, invasion and metastasis, and pro-osteoclastic factors.
- Published
- 2018
38. Abstract 4674: Targeting cancer stem-cells in aggressive variant prostate cancers
- Author
-
Rama Soundararajan, Paul Allegakoen, Petra den Hollander, Anurag Paranjape, Robiya Joseph, Sandhya Sundaram, Devarajan Karunagaran, Peter Shepherd, Nora Navone, Ana Aparicio, Sankar Maity, Bradley Broom, Christopher Logothetis, and Sendurai Mani
- Subjects
Cancer Research ,Oncology - Abstract
Aggressive Variant Prostate Cancers (AVPC) are lethal variants of the disease and usually occur in the setting of castration-resistant prostate cancer (CRPC). These neuroendocrine tumors are currently incurable, with most patients dying within 12-24 months of diagnosis. AVPCs are androgen-indifferent, and therefore do not respond to Androgen-Deprivation-Therapy (ADT), the mainstay treatment for prostate cancers. We recently discovered that these tumors are enriched with cancer cells with stem-cell properties (CSCs) that lack expression of androgen-receptor (AR) (PMID 26804168). Importantly, we observed that CSCs are generated when prostate cancer cells undergo a cell biological process called epithelial-to-mesenchymal-transition (or EMT). We also demonstrated that it is CSCs that facilitate neuroendocrine trans-differentiation, and promote drug-resistance via p38MAPK signaling. In this project, we aimed to inhibit EMT in patient-derived AVPC tumors (using SB203580, a p38MAPK inhibitor and anti-EMT drug), and identify clinically-relevant biological pathways upon EMT inhibition, using RNA-Seq analyses. We also aimed to develop a diagnostic CSC gene-expression score to identify/predict AVPC tumors. We tested the effect of SB203580 in two well-validated AR-negative PDX models of human AVPC (144-4 and 177-B). These models effectively recapitulate the heterogeneity and complex biology of AVPC. We first determined the appropriate dosing for SB203580 in these models that resulted in statistically significant target (p38MAPK) inhibition. Our data indicated that 10mg/kg SB significantly inhibits p38 activity (as judged by loss in MSK1 phosphorylation - MSK1 is a direct target of p38, and its phosphorylation is dependent on p38 activity). EMT inhibition also resulted in a significant reduction in the expression of SOX2, a known CSC marker for aggressive prostate cancers. In parallel, we observed a simultaneous up-regulation in expression of FOXA1, suggesting a shift to luminal epithelial phenotype upon EMT inhibition. We also developed a novel CSC gene expression score based on p38MAPK signaling components critical for EMT and neuroendocrine trans-differentiation in prostate cancer. We validated it in the Beltran dataset (PMID 26855148), wherein patients are categorized into CRPC-Adenocarcinoma and CRPC-Neuroendocrine type (aggressive tumors displaying extensive RB loss and increased expression of clinical neuroendocrine markers). We observed that our CSC score is highly represented in the CRPC-Neuro group, thus validating our rationale for anti-EMT therapy. This study thus highlights a potentially targetable signaling pathway for treatment of AVPC. Acknowledgements: We thank Dr. Mahajan (Washington University School of Medicine), Dr. Wistuba, Ms. Mino, Dr. Lin (UTMDACC) and Dr. Tang (Rosewell Park Cancer Institute) for their contributions, & UTMDACC Prostate Cancer SPORE for financial support (RS, SAM). Citation Format: Rama Soundararajan, Paul Allegakoen, Petra den Hollander, Anurag Paranjape, Robiya Joseph, Sandhya Sundaram, Devarajan Karunagaran, Peter Shepherd, Nora Navone, Ana Aparicio, Sankar Maity, Bradley Broom, Christopher Logothetis, Sendurai Mani. Targeting cancer stem-cells in aggressive variant prostate 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 4674.
- Published
- 2019
39. Targeting prostate cancer bone metastases
- Author
-
Shi-Ming Tu, Nora Navone, and Christopher Logothetis
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Bone disease ,business.industry ,Prostatic Neoplasms ,Bone Neoplasms ,medicine.disease ,Survival Analysis ,Metastasis ,Prostate cancer ,medicine.anatomical_structure ,Prostate ,Internal medicine ,Disease Progression ,Strontium Radioisotopes ,medicine ,Humans ,Adenocarcinoma ,Anthracyclines ,Prostate disease ,business ,Randomized Controlled Trials as Topic - Published
- 2003
40. The mechanism of growth-inhibitory effect of DOC-2/DAB2 in prostate cancer. Characterization of a novel GTPase-activating protein associated with N-terminal domain of DOC-2/DAB2
- Author
-
Zhi, Wang, Ching-Ping, Tseng, Rey-Chen, Pong, Hong, Chen, John D, McConnell, Nora, Navone, and Jer-Tsong, Hsieh
- Subjects
Male ,DNA, Complementary ,Base Sequence ,Sequence Homology, Amino Acid ,Tumor Suppressor Proteins ,GTPase-Activating Proteins ,Molecular Sequence Data ,Prostatic Neoplasms ,Proteins ,Adaptor Proteins, Vesicular Transport ,ras GTPase-Activating Proteins ,COS Cells ,Tumor Cells, Cultured ,Animals ,Humans ,Genes, Tumor Suppressor ,Amino Acid Sequence ,Cloning, Molecular ,Apoptosis Regulatory Proteins ,Cell Division ,Adaptor Proteins, Signal Transducing ,DNA Primers - Abstract
DOC-2/DAB2 is a member of the disable gene family with tumor-inhibitory activity. Its down-regulation is associated with several neoplasms, and serine phosphorylation of its N terminus modulates DOC-2/DAB2's inhibitory effect on AP-1 transcriptional activity. We describe the cloning of DIP1/2, a novel gene that interacts with the N-terminal domain of DOC-2/DAB2. DIP1/2 is a novel GTPase-activating protein containing a Ras GTPase-activating protein homology domain (N terminus) and two other unique domains (i.e. 10 proline repeats and leucine zipper). Interaction between DOC-2/DAB2 and DIP1/2 is detected in normal tissues such as the brain and prostate. Altered expression of these two proteins is often detected in prostate cancer cells. Indeed, the presence of DIP1/2 effectively blocks mitogen-induced gene expression and inhibits the growth of prostate cancer. Thus, DOC-2/DAB2 and DIP1/2 appear to represent a unique negative regulatory complex that maintains cell homeostasis.
- Published
- 2002
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.