27 results on '"Sepideh Vahid"'
Search Results
2. Supplementary Methods, Figure Legends from The Master Neural Transcription Factor BRN2 Is an Androgen Receptor–Suppressed Driver of Neuroendocrine Differentiation in Prostate Cancer
- Author
-
Amina Zoubeidi, Himisha Beltran, Yuzhuo Wang, Colin C. Collins, Mark A. Rubin, Dong Lin, Martin E. Gleave, Ladan Fazli, Alexander W. Wyatt, Fraser Johnson, Arkhjamil Angeles, Ka Mun Nip, Randy Jama, Hidetoshi Kuruma, Kirsi Ketola, Alastair Davies, Sepideh Vahid, Daksh Thaper, and Jennifer L. Bishop
- Abstract
Supplementary methods, figure legends, and primer lists.
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Table S1 from The Master Neural Transcription Factor BRN2 Is an Androgen Receptor–Suppressed Driver of Neuroendocrine Differentiation in Prostate Cancer
- Author
-
Amina Zoubeidi, Himisha Beltran, Yuzhuo Wang, Colin C. Collins, Mark A. Rubin, Dong Lin, Martin E. Gleave, Ladan Fazli, Alexander W. Wyatt, Fraser Johnson, Arkhjamil Angeles, Ka Mun Nip, Randy Jama, Hidetoshi Kuruma, Kirsi Ketola, Alastair Davies, Sepideh Vahid, Daksh Thaper, and Jennifer L. Bishop
- Abstract
RNAseq of ENZ-resistant cells.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Table S2 from The Master Neural Transcription Factor BRN2 Is an Androgen Receptor–Suppressed Driver of Neuroendocrine Differentiation in Prostate Cancer
- Author
-
Amina Zoubeidi, Himisha Beltran, Yuzhuo Wang, Colin C. Collins, Mark A. Rubin, Dong Lin, Martin E. Gleave, Ladan Fazli, Alexander W. Wyatt, Fraser Johnson, Arkhjamil Angeles, Ka Mun Nip, Randy Jama, Hidetoshi Kuruma, Kirsi Ketola, Alastair Davies, Sepideh Vahid, Daksh Thaper, and Jennifer L. Bishop
- Abstract
NEPC signature for GSEA analysis.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Table S3 from The Master Neural Transcription Factor BRN2 Is an Androgen Receptor–Suppressed Driver of Neuroendocrine Differentiation in Prostate Cancer
- Author
-
Amina Zoubeidi, Himisha Beltran, Yuzhuo Wang, Colin C. Collins, Mark A. Rubin, Dong Lin, Martin E. Gleave, Ladan Fazli, Alexander W. Wyatt, Fraser Johnson, Arkhjamil Angeles, Ka Mun Nip, Randy Jama, Hidetoshi Kuruma, Kirsi Ketola, Alastair Davies, Sepideh Vahid, Daksh Thaper, and Jennifer L. Bishop
- Abstract
Gene Profiling data of CRPC cells with BRN2 over-expression.
- Published
- 2023
- Full Text
- View/download PDF
6. Discovery and characterization of a first-in-field transcription factor BRN2 inhibitor for the treatment of neuroendocrine prostate cancer
- Author
-
Daksh Thaper, Ravi Munuganti, Adeleke Aguda, Soojin Kim, Shengyu Ku, Olena Sivak, Sahil Kumar, Sepideh Vahid, Dwaipayan Ganguli, Himisha Beltran, Colm Morrissey, Eva Corey, and Amina Zoubeidi
- Abstract
The increased incidence of treatment-emergent neuroendocrine prostate cancer (NEPC) is particularly alarming as this diagnosis is associated with poor prognosis. Despite initial responses to platinum-based chemotherapy, relapses are common and there is no effective second line therapy for NEPC. We previously identified that neuronal transcription factor BRN2 (POU3F2) is a potent driver of neuroendocrine differentiation and an attractive target for NEPC. Utilizing a combination of in silico modeling and X-ray crystallography followed by structure-based lead optimization, we have developed the first potent, specific and orally bioavailable BRN2 inhibitor (B18-94), which inhibits the interaction between BRN2 and DNA. This loss of BRN2 on the chromatin drastically reduces its transcriptional output resulting in downregulation of several known targets in NEPC such as SOX2, ASCL1 and PEG10. Additionally, B18-94 reduces specifically cell proliferation specifically in multiple NEPC models with no effect on adenocarcinoma and other BRN2 negative prostate cancer models. Importantly, the consistency in the transcriptomic changes driven by B18-94 and or CRISPR/Cas9 mediated BRN2 knockout confirmed the on-target specificity, with both methods of BRN2 inhibition downregulating pathways involved in cellular plasticity and proliferation. Finally, we have demonstrated that B18-94, the first-in-field POU-domain transcription factor inhibitor, significantly reduced tumor growth in several NEPC xenograft models with no observable toxicity, suggesting potential for therapeutic intervention of NEPC.
- Published
- 2022
- Full Text
- View/download PDF
7. Targeting Lyn regulates Snail family shuttling and inhibits metastasis
- Author
-
Ka Mun Nip, Kirsi Ketola, Amina Zoubeidi, Cheryl Y. Gregory-Evans, Kenneth W. Harder, Sebastian Frees, X Shan, Sepideh Vahid, Igor Moskalev, Jennifer L. Bishop, Morgan E. Roberts, and Daksh Thaper
- Subjects
0301 basic medicine ,Cancer Research ,Epithelial-Mesenchymal Transition ,Biology ,Molecular oncology ,Mice ,03 medical and health sciences ,Growth factor receptor ,Downregulation and upregulation ,LYN ,Cell Line, Tumor ,Neoplasms ,Genetics ,Animals ,Humans ,Molecular Targeted Therapy ,Neoplasm Metastasis ,RNA, Small Interfering ,Cell adhesion ,Molecular Biology ,Transcription factor ,Cell cycle ,Xenograft Model Antitumor Assays ,Cell biology ,Gene Expression Regulation, Neoplastic ,Protein Transport ,src-Family Kinases ,030104 developmental biology ,Tumor progression ,Snail Family Transcription Factors - Abstract
The acquisition of an invasive phenotype by epithelial cells occurs through a loss of cellular adhesion and polarity, heralding a multistep process that leads to metastatic dissemination. Since its characterization in 1995, epithelial-mesenchymal transition (EMT) has been closely linked to the metastatic process. As a defining aspect of EMT, loss of cell adhesion through downregulation of E-cadherin is carried out by several transcriptional repressors; key among them the SNAI family of transcription factors. Here we identify for the first time that Lyn kinase functions as a key modulator of SNAI family protein localization and stability through control of the Vav-Rac1-PAK1 (Vav-Rac1-p21-activated kinase) pathway. Accordingly, targeting Lyn in vitro reduces EMT and in vivo reduces metastasis of primary tumors. We also demonstrate the clinical relevance of targeting Lyn as a key player controlling EMT; patient samples across many cancers revealed a strong negative correlation between Lyn and E-cadherin, and high Lyn expression in metastatic tumors as well as metastasis-prone primary tumors. This work reveals a novel pancancer mechanism of Lyn-dependent control of EMT and further underscores the role of this kinase in tumor progression.
- Published
- 2017
- Full Text
- View/download PDF
8. Neural Transcription Factors in Disease Progression
- Author
-
Daksh, Thaper, Sepideh, Vahid, and Amina, Zoubeidi
- Subjects
Gene Expression Regulation, Neoplastic ,Male ,Receptors, Androgen ,Disease Progression ,Humans ,Prostatic Neoplasms ,Cell Cycle Proteins ,Carcinoma, Neuroendocrine ,Transcription Factors - Abstract
Progression to the malignant state is fundamentally dependent on transcriptional regulation in cancer cells. Optimum abundance of cell cycle proteins, angiogenesis factors, immune evasion markers, etc. is needed for proliferation, metastasis or resistance to treatment. Therefore, dysregulation of transcription factors can compromise the normal prostate transcriptional network and contribute to malignant disease progression.The androgen receptor (AR) is considered to be a key transcription factor in prostate cancer (PCa) development and progression. Consequently, androgen pathway inhibitors (APIs) are currently the mainstay in PCa treatment, especially in castration-resistant prostate cancer (CRPC). However, emerging evidence suggests that with increased administration of potent APIs, prostate cancer can progress to a highly aggressive disease that morphologically resembles small cell carcinoma, which is referred to as neuroendocrine prostate cancer (NEPC), treatment-induced or treatment-emergent small cell prostate cancer. This chapter will review how neuronal transcription factors play a part in inducing a plastic stage in prostate cancer cells that eventually progresses to a more aggressive state such as NEPC.
- Published
- 2020
9. The Master Neural Transcription Factor BRN2 Is an Androgen Receptor–Suppressed Driver of Neuroendocrine Differentiation in Prostate Cancer
- Author
-
Yuzhuo Wang, Jennifer L. Bishop, Randy Jama, Alexander W. Wyatt, Ka Mun Nip, Mark A. Rubin, Kirsi Ketola, Amina Zoubeidi, Dong Lin, Daksh Thaper, Sepideh Vahid, Himisha Beltran, Arkhjamil Angeles, Martin E. Gleave, Alastair H. Davies, Hidetoshi Kuruma, Ladan Fazli, Colin Collins, and Fraser Johnson
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Transcription, Genetic ,Cellular differentiation ,Biology ,Neuroendocrine differentiation ,Gene Knockout Techniques ,Mice ,03 medical and health sciences ,Prostate cancer ,SOX2 ,Cell Line, Tumor ,Internal medicine ,Nitriles ,Phenylthiohydantoin ,medicine ,Animals ,Humans ,Transcription factor ,Homeodomain Proteins ,Regulation of gene expression ,SOXB1 Transcription Factors ,Prostatic Neoplasms ,Cell Differentiation ,medicine.disease ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Androgen receptor ,030104 developmental biology ,Endocrinology ,Oncology ,Drug Resistance, Neoplasm ,Receptors, Androgen ,Benzamides ,POU Domain Factors ,Disease Progression ,Cancer research ,Adenocarcinoma ,Neoplasm Transplantation - Abstract
Mechanisms controlling the emergence of lethal neuroendocrine prostate cancer (NEPC), especially those that are consequences of treatment-induced suppression of the androgen receptor (AR), remain elusive. Using a unique model of AR pathway inhibitor–resistant prostate cancer, we identified AR-dependent control of the neural transcription factor BRN2 (encoded by POU3F2) as a major driver of NEPC and aggressive tumor growth, both in vitro and in vivo. Mechanistic studies showed that AR directly suppresses BRN2 transcription, which is required for NEPC, and BRN2-dependent regulation of the NEPC marker SOX2. Underscoring its inverse correlation with classic AR activity in clinical samples, BRN2 expression was highest in NEPC tumors and was significantly increased in castration-resistant prostate cancer compared with adenocarcinoma, especially in patients with low serum PSA. These data reveal a novel mechanism of AR-dependent control of NEPC and suggest that targeting BRN2 is a strategy to treat or prevent neuroendocrine differentiation in prostate tumors.Significance: Understanding the contribution of the AR to the emergence of highly lethal, drug-resistant NEPC is critical for better implementation of current standard-of-care therapies and novel drug design. Our first-in-field data underscore the consequences of potent AR inhibition in prostate tumors, revealing a novel mechanism of AR-dependent control of neuroendocrine differentiation, and uncover BRN2 as a potential therapeutic target to prevent emergence of NEPC. Cancer Discov; 7(1); 54–71. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 1
- Published
- 2017
- Full Text
- View/download PDF
10. Abstract 3083: Selective inhibition of transcription factor BRN2 as a treatment strategy for Small Cell Prostate Cancer
- Author
-
Amina Zoubeidi, Soo Jin Kim, Shungyu Ku, Olena Sivak, Sahil Kumar, Daksh Thaper, Shaghayegh Nouruzi, Adeleke Aguda, Ravi Shashi Nayana Munuganti, Dwaiyapan Ganguli, Eva Corey, Colm Morrissey, Himisha Beltran, and Sepideh Vahid
- Subjects
Cancer Research ,Cell growth ,Cancer ,Biology ,medicine.disease ,Neuroendocrine differentiation ,Androgen receptor ,chemistry.chemical_compound ,Prostate cancer ,Oncology ,chemistry ,SOX2 ,Cancer research ,medicine ,Enzalutamide ,Transcription factor - Abstract
Introduction: Resistance to newly developed androgen receptor pathway inhibitors (ARPIs), such as Enzalutamide (ENZ), rapidly emerges. In particular, a subset of patients who relapse following ARPI therapy their dependence on AR signaling and emerge with neuroendocrine features. These tumors, termed treatment induced small-cell prostate cancer (t-SCPC) or neuroendocrine prostate cancer (t-NEPC), carry an extremely poor prognosis and, to date, treatment remains decades old cytotoxic chemotherapies. Previously, our group identified the neural transcription factor BRN2 as a major clinically relevant driver of SCPC and now report that targeting BRN2 is a promising strategy to prevent neuroendocrine differentiation or treat NEPC.Methods/Results: In silico screening of small molecules was conducted on a model of BRN2 which was later validated with the first-in-field crystal structure of BRN2 DNA binding domain. On the basis of the model, several small molecules were identified that showed direct binding to BRN2 and inhibited its activity. Pharmacokinetic studies measured stability and bioavailability of med-chem optimized lead compound (BRN2i) that significantly reduced tumor growth in multiple xenograft models with no measurable side-effects. In silico modeling predicted a 7Å “closing” in the DBD once it was bound to BRN2i, this shift translated to reduced interaction with DNA by chromatin fractionation and ChIP-seq, thus confirming the mode of action for BRN2i is through loss of DNA binding. Loss of BRN2 binding reduced cell proliferation in tSCPC cell line 42DENZR, de novo SCPC cell line NCI-H660 and NEPC organoids as well as downregulated several known targets like EZH2, ASCL1, SOX2 and PEG10. Down-regulation of these target genes was also measured in the xenograft tumors, confirming on target effect in vivo. Moreover, the specificity of BRN2i was validated with CRISPR/Cas9 mediated knockout of BRN2 with downstream mRNA expression and phenotypic changes. Conclusion: The described work aims to lay the pre-clinical foundation for the integration of BRN2 targeted therapies into the treatment landscape to improve survival for patients suffering from small-cell neuroendocrine prostate cancer. Citation Format: Daksh Thaper, Ravi Munuganti, Adeleke Aguda, Soojin Kim, Shungyu Ku, Sahil Kumar, Sepideh Vahid, Shaghayegh Nouruzi, Olena Sivak, Dwaiyapan Ganguli, Colm Morrissey, Eva Corey, Himisha Beltran, Amina Zoubeidi. Selective inhibition of transcription factor BRN2 as a treatment strategy for Small Cell Prostate Cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 3083.
- Published
- 2020
- Full Text
- View/download PDF
11. Galiellalactone inhibits the STAT3/AR signaling axis and suppresses Enzalutamide-resistant Prostate Cancer
- Author
-
Shaghayegh Nouruzi, Ramandeep Kaur, Sepideh Vahid, Daksh Thaper, Sahil Kumar, Amina Zoubeidi, Jennifer L. Bishop, and Martin Johansson
- Subjects
Male ,STAT3 Transcription Factor ,0301 basic medicine ,lcsh:Medicine ,Mice, Nude ,Apoptosis ,urologic and male genital diseases ,Article ,Lactones ,Mice ,03 medical and health sciences ,Prostate cancer ,chemistry.chemical_compound ,0302 clinical medicine ,Cyclin D1 ,Nitriles ,Phenylthiohydantoin ,LNCaP ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Enzalutamide ,lcsh:Science ,STAT3 ,Transcription factor ,Cell Proliferation ,Multidisciplinary ,biology ,Cell growth ,lcsh:R ,medicine.disease ,Xenograft Model Antitumor Assays ,3. Good health ,Androgen receptor ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,chemistry ,Drug Resistance, Neoplasm ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Benzamides ,Cancer research ,biology.protein ,lcsh:Q - Abstract
Most prostate cancer patients will progress to a castration-resistant state (CRPC) after androgen ablation therapy and despite the development of new potent anti-androgens, like enzalutamide (ENZ), which prolong survival in CRPC, ENZ-resistance (ENZR) rapidly occurs. Re-activation of the androgen receptor (AR) is a major mechanism of resistance. Interrogating our in vivo derived ENZR model, we discovered that transcription factor STAT3 not only displayed increased nuclear localization but also bound to and facilitated AR activity. We observed increased STAT3 S727 phosphorylation in ENZR cells, which has been previously reported to facilitate AR binding. Strikingly, ENZR cells were more sensitive to inhibition with STAT3 DNA-binding inhibitor galiellalactone (GPA500) compared to CRPC cells. Treatment with GPA500 suppressed AR activity and significantly reduced expression of Cyclin D1, thus reducing cell cycle progression into S phase and hindering cell proliferation. In vivo, GPA500 reduced tumor volume and serum PSA in ENZR xenografts. Lastly, the combination of ENZ and GPA500 was additive in the inhibition of AR activity and proliferation in LNCaP and CRPC cells, providing rationale for combination therapy. Overall, these results suggest that STAT3 inhibition is a rational therapeutic approach for ENZR prostate cancer, and could be valuable in CRPC in combination with ENZ.
- Published
- 2018
- Full Text
- View/download PDF
12. Hsp27 regulates EGF/β-catenin mediated epithelial to mesenchymal transition in prostate cancer
- Author
-
Ka Mun Nip, Masaki Shiota, Sepideh Vahid, Daksh Thaper, Martin E. Gleave, Devon Heroux, Jennifer L. Bishop, Amina Zoubeidi, and Thomas Cordonnier
- Subjects
Cancer Research ,animal structures ,biology ,Cell migration ,Vimentin ,urologic and male genital diseases ,medicine.disease ,Metastasis ,Oncology ,Epidermal growth factor ,Catenin ,embryonic structures ,Cancer cell ,medicine ,Cancer research ,biology.protein ,Epithelial–mesenchymal transition ,Signal transduction - Abstract
Increased expression of the molecular chaperone Hsp27 is associated with the progression of prostate cancer (PCa) to castration-resistant disease, which is lethal due to metastatic spread of the prostate tumor. Metastasis requires epithelial to mesenchymal transition (EMT), which endows cancer cells with the ability to disseminate from the primary tumor and colonize new tissue sites. A wide variety of secreted factors promote EMT, and while overexpression and constitutive activation of epidermal growth factor (EGF) signaling is associated with poor prognosis of PCa, a precise role of EGF in PCa progression to metastasis remains unclear. Here, we show that Hsp27 is required for EGF-induced cell migration, invasion and MMPs activity as well as the expression of EMT markers including Fibronectin, Vimentin and Slug with concomitant decrease of E-cadherin. Mechanistically, we found that Hsp27 is required for EGF-induced AKT and GSK3β phosphorylation and β-catenin nuclear translocation. Moreover, silencing Hsp27 decreases EGF dependent phosphorylation of β-catenin on tyrosine 142 and 654, enhances β-catenin ubiquitination and degradation, prevents β-catenin nuclear translocation and binding to the Slug promoter. These data suggest that Hsp27 is required for EGF-mediated EMT via modulation of the β-catenin/Slug signaling pathway. Together, our findings underscore the importance of Hsp27 in EGF induced EMT in PCa and highlight the use of Hsp27 knockdown as a useful strategy for patients with advanced disease.
- Published
- 2014
- Full Text
- View/download PDF
13. Abstract 1295: First-in-field small molecule inhibitors targeting BRN2 as a therapeutic strategy for small cell prostate cancer
- Author
-
Daksh Thaper, Ravi Munuganti, Shaghayegh Nouruzi, Sahil Kumar, Soojin Kim, Olena Sivak, Adeleke Aguda, Dwaipayan Ganguli, Sepideh Vahid, Loredana Puca, Himisha Beltran, and Amina Zoubeidi
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Resistance to newly developed androgen receptor pathway inhibitors (ARPIs), such as Enzalutamide (ENZ), rapidly emerges and patients generally die within two years. In particular, a subset of patients who relapse following ARPI therapy exhibit lineage switching whereby tumours shed their dependence on AR signaling and emerge with neuroendocrine features. These tumours, termed treatment induced neuroendocrine prostate cancer (t-NEPC), carry an extremely poor prognosis and, to date, treatment remains decades old cytotoxic chemotherapy which carries a short-lived response at the cost of significant toxicity. Therefore, targeted therapies for this deadly disease are desperately needed. Thus, the need to develop targeted treatments for this devastating disease is of paramount importance. Recently our group identified the neural transcription factor BRN2 as a major clinically relevant driver of NEPC and aggressive tumor growth, both in vitro and in vivo, suggesting targeting BRN2 is a promising strategy to prevent neuroendocrine differentiation or treat NEPC. Methods: Study the effects of BRN2 inhibition using siRNA, small molecule inhibitors and CRISPR K/O models. Results: Inhibition of BRN2 by siRNA and by CRISPR/Cas9 knockout drastically reduced cell proliferation in 42DENZR (NEPC) cell lines. This data was re-capitulated in human NEPC NCI-H660 cells. Loss of BRN2 initiated drastic epigenetic changes in NEPC cell lines as well as in G1 arrest through up-regulation of CDKN1A/1B. This was confirmed using our first in field BRN2 inhibitors. Targeting BRN2 also lead to downregulation several known targets in NEPC like EZH2, AURKA, SOX2 and Peg10. Treatment with BRN2i reduced recruitment of BRN2 to the chromatin by approximately 93% within 16 hours. Moreover, these BRN2 inhibitors displayed adequate pharmacokinetic properties and reduced NEPC proliferation in vivo. Conclusion: No therapies exist for highly lethal NEPC. Hence, the described work aims to verify BRN2, a central driver of NEPC, and lay the pre-clinical foundation for the integration of targeted therapies into the treatment landscape to improve survival and quality of life for patients suffering from deadly form of prostate cancer. Citation Format: Daksh Thaper, Ravi Munuganti, Shaghayegh Nouruzi, Sahil Kumar, Soojin Kim, Olena Sivak, Adeleke Aguda, Dwaipayan Ganguli, Sepideh Vahid, Loredana Puca, Himisha Beltran, Amina Zoubeidi. First-in-field small molecule inhibitors targeting BRN2 as a therapeutic strategy for small cell 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 1295.
- Published
- 2019
- Full Text
- View/download PDF
14. First-in-field small molecule inhibitors targeting BRN2 as a therapeutic strategy for small cell prostate cancer
- Author
-
Amina Zoubeidi, Soojin Kim, Olena Sivak, Ravi Shashi Nayana Munuganti, Adeleke H Aguda, Daksh Thaper, Sepideh Vahid, Himisha Beltran, Loredana Puca, Sahil Kumar, and Shaghayegh Nouruzi
- Subjects
Cancer Research ,business.industry ,Cell ,medicine.disease ,Small molecule ,Androgen receptor ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Enzalutamide ,business ,030215 immunology ,Therapeutic strategy - Abstract
260 Background: Resistance to newly developed androgen receptor pathway inhibitors (ARPIs), such as Enzalutamide (ENZ), rapidly emerges and patients generally die within two years. In particular, a subset of patients who relapse following ARPI therapy exhibit lineage switching whereby tumours shed their dependence on AR signaling and emerge with neuroendocrine features. These tumours, termed treatment induced neuroendocrine prostate cancer (t-NEPC), carry an extremely poor prognosis and, to date, treatment remains decades old cytotoxic chemotherapies; therefore, targeted therapies are desperately needed. Recently our group identified the neural transcription factor BRN2 as a major clinically relevant driver of NEPC and aggressive tumor growth, both in vitro and in vivo, suggesting targeting BRN2 is a promising strategy to prevent neuroendocrine differentiation or treat NEPC. Methods: Study the effects of BRN2 inhibition using siRNA, small molecule inhibitors (BRN2i) and CRISPR K/O models. The efficacy of the small molecules was examined using reporter assays, florescence polarization assays, Biolayer Interferometry, DARTS, chromatin fractionation, RNA-seq and ChIP-seq. Pharmacokinetic studies measured stability and bioavailability of the molecules and in-vivo efficacy is to be measured in NCI-H660 xenograft model. Results: Inhibition of BRN2 drastically reduced cell proliferation in NEPC cell lines 42DENZR and NCI-H660 cell lines. Targeting BRN2 with our first-in-field small molecule inhibitors lead to downregulation several known targets in NEPC like EZH2, ASCL1, SOX2 and PEG10. Treatment with BRN2i reduced recruitment of BRN2 to the chromatin by approximately 93% within 16 hours. Moreover, these BRN2 inhibitors displayed adequate pharmacokinetic properties and reduced NEPC proliferation in vivo. Conclusions: No therapies exist for highly lethal NEPC. Hence, the described work aims to lay the pre-clinical foundation for the integration of BRN2 targeted therapies into the treatment landscape to improve survival for patients suffering from small cell prostate cancer.
- Published
- 2019
- Full Text
- View/download PDF
15. Immunogenomic landscape of neuroendocrine small cell prostate cancer
- Author
-
Martin E. Gleave, Christian Kollmannsberger, Bernhard J. Eigl, Amina Zoubeidi, Lucia Nappi, Alexander W. Wyatt, Ladan Fazli, Sepideh Vahid, Kim N. Chi, and Claudia Kesch
- Subjects
Cisplatin ,Cancer Research ,Poor prognosis ,business.industry ,medicine.medical_treatment ,Cell ,medicine.disease ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Hormone therapy ,business ,030215 immunology ,medicine.drug - Abstract
217 Background: Neuroendocrine small cell prostate cancer (NEPC) is a lethal variant of prostate cancer (PCa) unresponsive to hormone therapy and associated with poor prognosis. Cisplatin induces short-lived responses and therefore alternative novel therapeutic options are urgently needed. Methods: Prostate specimens from radical prostatectomy or transurethral resection (benign prostate specimens n = 4, primary untreated or neoadjuvant hormone-treated adenocarcinoma n = 30, castrate-resistant prostate cancer-CRPC n = 38 and NEPC n = 16) were evaluated for PD-L1 (SpringBio, M4420), AR, chromogranin A, synaptophysin, NSE and CD56 expression by immunohistochemistry (IHC). Archival tissue from liver, lymph nodes and prostate from 30 additional patients with de novo and treatment emergent NEPC were analyzed for PD-L1 expression by IHC. The intensity was assessed as percentage of positive cells / mm2 of tissue. Targeted and whole exome sequencing of the high-density tumor areas were performed and correlated to the PD-L1 status (PD-L1+ve: > 1% of positive cells). OS was calculated from the date of diagnosis of NEPC to death. We set out to define PD-L1 expression and immunogenomic characteristics of NEPC. Results: PD-L1 was expressed in 0%, 5%, 10% and 41% of benign, adenocarcinoma, CRPC and NEPC specimens, respectively. The PD-L1 expression intensity was significantly higher in patients with NEPC (mean: 40%, range: 5-100%) compared to benign, adenocarcinoma and CRPC samples (mean: 0%, 2% and 8%, respectively, P < 0.0001). There was a higher prevalence of biallelic DNA Repair Defects (DRD) in the PD-L1+ve vs PD-L1-ve patients (65% vs 0%, P = 0.005). The median OS of the NEPC patients was 8.5 months vs 10.5 months in PD-L1+ve vs PD-L1-ve tumors (HR 1.24, 95% CI: 0.59-2.75, p = 0.55). Conclusions: NEPC have greater PD-L1 expression than adenoCa and CRPC. Biallelic DRD was exclusively observed in PD-L1+ve patients. Since PD-L1 expression and DRD have been associated to response to PARP and PD1/PD-L1 inhibitors in prostate and other cancers, further studies evaluating the activity of those agents in NEPC patients are warranted.
- Published
- 2019
- Full Text
- View/download PDF
16. Chaperoning the Cancer: The Proteostatic Functions of the Heat Shock Proteins in Cancer
- Author
-
Amina Zoubeidi, Daksh Thaper, and Sepideh Vahid
- Subjects
0301 basic medicine ,Cancer Research ,Cell Survival ,Antineoplastic Agents ,Protein degradation ,Bioinformatics ,Patents as Topic ,03 medical and health sciences ,Hsp27 ,Heat shock protein ,Neoplasms ,Drug Discovery ,medicine ,Animals ,Homeostasis ,Humans ,Pharmacology (medical) ,Proteostasis Deficiencies ,Heat-Shock Proteins ,Clinical Trials as Topic ,biology ,Cancer ,General Medicine ,medicine.disease ,Hsp90 ,Cell biology ,Hsp70 ,030104 developmental biology ,Proteostasis ,Oncology ,Cancer cell ,biology.protein ,Molecular Chaperones - Abstract
Background: Protein homeostasis (proteostasis) is vital for the survival of cells in physiological and pathological conditions. Particularly, cancer cells are in constant state of cellular stress due to rapid proliferation and decreased quality control in proteosynthesis and therefore, are exceedingly dependent on the homeostasis pathways. Among the complex biological mechanisms regulating proteostasis are the highly conserved molecular chaperones, heat shock proteins (HSPs). HSPs assist cell survival by catalysing the proper folding of proteins, modulation of the apoptotic machinery and finally regulating the protein degradation machinery, providing either the stability or the degradation of selected proteins under stress conditions. Inevitably, HSPs are upregulated in malignancies and participate in different hallmarks of cancer, with indispensable roles in the onset and progression of the disease. Moreover, high levels of HSPs contribute to poor prognosis and treatment resistance in various cancers. Therefore these molecular chaperones present as attractive targets for anti-cancer therapy. Objective: This review describes how HSPs regulate different hallmarks of cancer and provides an overview on the most relevant patents which have recently appeared in the literature. Methods: The patents were extracted from Google Patents (2012-2016) while the clinical trial results were mined from www.clinicaltrial.gov. Results and Conclusion: Review of literature shows that the proteostatic functions of HSPs can modify different hallmarks of cancer. Moreover, targeting HSPs (notably HSP27, HSP70 and HSP90) exhibited positive results in clinical trials so far. However, more studies should be designed to optimize the efficacy of mono or combination therapy in various malignancies.
- Published
- 2016
17. Molecular chaperone Hsp27 regulates the Hippo tumor suppressor pathway in cancer
- Author
-
Jennifer L. Bishop, Amina Zoubeidi, Kate Gibson, Daksh Thaper, and Sepideh Vahid
- Subjects
Male ,0301 basic medicine ,Cytoplasm ,endocrine system ,MST1 ,Lung Neoplasms ,animal structures ,HSP27 Heat-Shock Proteins ,Breast Neoplasms ,Protein Serine-Threonine Kinases ,Article ,03 medical and health sciences ,Hsp27 ,Cell Line, Tumor ,Neoplasms ,Humans ,Hippo Signaling Pathway ,Phosphorylation ,Heat-Shock Proteins ,Adaptor Proteins, Signal Transducing ,Regulation of gene expression ,Hippo signaling pathway ,Multidisciplinary ,biology ,Kinase ,Intracellular Signaling Peptides and Proteins ,Prostatic Neoplasms ,YAP-Signaling Proteins ,Genomics ,Gene signature ,Phosphoproteins ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,14-3-3 Proteins ,A549 Cells ,Transcriptional Coactivator with PDZ-Binding Motif Proteins ,Trans-Activators ,Cancer research ,biology.protein ,Female ,Signal transduction ,Molecular Chaperones ,Signal Transduction ,Transcription Factors - Abstract
Heat shock protein 27 (Hsp27) is a molecular chaperone highly expressed in aggressive cancers, where it is involved in numerous pro-tumorigenic signaling pathways. Using functional genomics we identified for the first time that Hsp27 regulates the gene signature of transcriptional co-activators YAP and TAZ, which are negatively regulated by the Hippo Tumor Suppressor pathway. The Hippo pathway inactivates YAP by phosphorylating and increasing its cytoplasmic retention with the 14.3.3 proteins. Gain and loss of function experiments in prostate, breast and lung cancer cells showed that Hsp27 knockdown induced YAP phosphorylation and cytoplasmic localization while overexpression of Hsp27 displayed opposite results. Mechanistically, Hsp27 regulates the Hippo pathway by accelerating the proteasomal degradation of ubiquitinated MST1, the core Hippo kinase, resulting in reduced phosphorylation/activity of LATS1 and MOB1, its downstream effectors. Importantly, our in vitro results were supported by data from human tumors; clinically, high expression of Hsp27 in prostate tumors is correlated with increased expression of YAP gene signature and reduced phosphorylation of YAP in lung and invasive breast cancer clinical samples. This study reveals for the first time a link between Hsp27 and the Hippo cascade, providing a novel mechanism of deregulation of this tumor suppressor pathway across multiple cancers.
- Published
- 2016
- Full Text
- View/download PDF
18. A NEW PRE-COLUMN DERIVATIZATION METHOD FOR DETERMINATION OF NITRITE AND NITRATE IN HUMAN PLASMA BY HPLC
- Author
-
Mohamadhosein Salehi Sormaghi, Sepideh Vahid, Simin Dashti-Khavidaki, Mohsen Amini, and Farrokhlegha Ahmadi
- Subjects
Analyte ,Chromatography ,Clinical Biochemistry ,Pharmaceutical Science ,Reversed-phase chromatography ,Biochemistry ,Analytical Chemistry ,chemistry.chemical_compound ,Nitrate ,chemistry ,Griess test ,Sample preparation ,Nitrite ,Sodium nitrite ,Derivatization - Abstract
Nitrite and nitrate are metabolites of nitric oxide (NO) which its fundamental role in biological systems is emphasized. Therefore, accurate measurement of these ions is profoundly important. Here, a simple, accurate, and sensitive method based on reversed phase liquid chromatography is developed. This HPLC technique includes pre-column derivatization with Griess reagents and detection at 540 nm. Sample preparation is easy and involves liquid-liquid back extraction which purifies the analyte and improves sensitivity. Lower limit of quantification for nitrite was found to be 66.7 nM which provides enough sensitivity to determine nitrite in many biological samples. Furthermore, reduction of nitrate with vanadium chloride (III) was optimized. An internal standard was used to enhance method's repeatability, making the correlation variations of intra and inter-day assays below 8.77% with less than 17.00% error at all concentrations. The method was linear in the range of 0.1–50 µM for sodium nitrite and 1–500 µ...
- Published
- 2012
- Full Text
- View/download PDF
19. Abstract 1931: Targeting master neuronal transcription factor BRN2 in neuroendocrine prostate cancer
- Author
-
Ravi Sn Munuganti, Sepideh Vahid, Soojin Kim, Shaghayegh Norouzi, Kriti Singh, Daksh Thaper, Jennifer L. Bishop, Sahil Kumar, and Amina Zoubeidi
- Subjects
Cancer Research ,Prostate cancer ,Oncology ,business.industry ,Cancer research ,Medicine ,business ,medicine.disease ,Transcription factor - Abstract
Introduction: Resistance to newly developed androgen receptor pathway inhibitors (ARPIs), such as Enzalutamide (ENZ), rapidly emerges and patients generally die within two years. In particular, a subset of patients who relapse following ARPI therapy exhibit lineage switching whereby tumours shed their dependence on AR signaling and emerge with neuroendocrine features. These tumours, termed treatment induced neuroendocrine prostate cancer (t-NEPC), carry an extremely poor prognosis and, to date, treatment remains decades old cytotoxic chemotherapy which carries a short-lived response at the cost of significant toxicity. Therefore, targeted therapies for this deadly disease are desperately needed. Thus, the need to develop targeted treatments for this devastating disease is of paramount importance. Recently our group identified the neural transcription factor BRN2 as a major clinically relevant driver of NEPC and aggressive tumor growth, both in vitro and in vivo, suggesting targeting BRN2 is a promising strategy to prevent neuroendocrine differentiation or treat NEPC. Methods: Study the effects of BRN2 inhibition using siRNA and CRISPR K/O models. Results: Inhibition of BRN2 by siRNA and by CRISPR/Cas9 knockout drastically reduced cell proliferation in 42DENZR (NEPC) cell lines. This data was re-capitulated in human NEPC NCI-H660 cells. Loss of BRN2 initiated drastic epigenetic changes in NEPC cell lines as well as in G1 arrest through up-regulation of CDKN1A/1B. This was confirmed using our first in field BRN2 inhibitors. Targeting BRN2 also lead to downregulation several known targets in NEPC like EZH2, AURKA, SOX2 and Peg10. Conclusion: No therapies exist for highly lethal NEPC. Hence, the described work aims to verify BRN2 as a central driver of NEPC, and lay the pre-clinical foundation for the integration of targeted therapies into the treatment landscape to improve survival and quality of life for patients suffering from deadly form of prostate cancer. Citation Format: Daksh Thaper, Ravi Munuganti, Shaghayegh Norouzi, Sahil Kumar, Soojin Kim, Kriti Singh, Sepideh Vahid, Jennifer Bishop, Amina Zoubeidi. Targeting master neuronal transcription factor BRN2 in neuroendocrine prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1931.
- Published
- 2018
- Full Text
- View/download PDF
20. Concomitant chemopreventive and antibacterial effects of some Iranian plants from the genus Cousinia (Asteraceae)
- Author
-
Farshid Saadat, Ahmad Reza Shahverdi, Mohammad Reza Khorramizadeh, Ahmad Ghahraman, Farideh Attar, and Sepideh Vahid
- Subjects
Cousinia ,biology ,Traditional medicine ,linhagem celular WEHI 164 ,citotoxicidade ,lcsh:RS1-441 ,zymography ,IC50 ,Asteraceae ,biology.organism_classification ,lcsh:Pharmacy and materia medica ,antibacterial ,Cell culture ,Botany ,cytotoxicity ,antibacteriano ,Zymography ,zimografia ,Agar diffusion test ,General Pharmacology, Toxicology and Pharmaceutics ,Cytotoxicity ,Antibacterial activity ,WEHI 164 cell line - Abstract
During the past several years, various species of Cousinia (Asteraceae) have been authenticated in Iran. However, data concerning their biological activities remain limited. The main purpose of this research was to assess potential cytotoxicity and matrix metalloproteinases (MMP) inhibitory effects of seven ethanol extracts of Cousinia using a cell line model (Fibrosarcoma-WEHI 164). We further investigated the antibacterial activity of these Cousinia ethanol extracts, using disk diffusion method. Among the ethanol extracts, the total extract of C. sulabadensis elicited significant inhibition of MMP activity in a dose-response fashion (49.2 ± 0.51, p < 0.05). However, this extract exhibited the lowest cytotoxicity effect at all tested concentrations. The concentration necessary to produce a 50% cell death rate (IC50) with C. shulabadensis was 304.5 ± 0.61 µg/mL. The calculated IC50 for cytotoxicity of the other Cousinia species extracts ranged between 18.4 ± 0.59 to 87.9 ± 0.58 µg/mL. The highest antibacterial activity was observed for the total extract of Cousinia phyllocephala. In conclusion, this study supports that Cousinia species display a remarkable inhibition of matrix metalloproteinases activity. The concomitant MMP-inhibitory and low cytotoxicity effects observed in C. sulabadensis might coin this extract for future potential anti-invasive herbal medicine studies.
- Published
- 2007
- Full Text
- View/download PDF
21. Lyn tyrosine kinase regulates androgen receptor expression and activity in castrate-resistant prostate cancer
- Author
-
Sepideh Vahid, Kilian M. Gust, Michael E. Cox, Jennifer L. Bishop, Paul Toren, Amina Zoubeidi, Ladan Fazli, Francois Lamoureux, Ka Mun Nip, Daksh Thaper, Anousheh Zardan, and Eliana Beraldi
- Subjects
Cancer Research ,Biology ,medicine.disease ,urologic and male genital diseases ,Hsp90 ,In vitro ,Androgen receptor ,Prostate cancer ,Downregulation and upregulation ,Ubiquitin ,LYN ,Cancer research ,biology.protein ,medicine ,Original Article ,Molecular Biology ,Tyrosine kinase - Abstract
Castrate-resistant prostate cancer (CRPC) progression is a complex process by which prostate cells acquire the ability to survive and proliferate in the absence or under very low levels of androgens. Most CRPC tumors continue to express the androgen receptor (AR) as well as androgen-responsive genes owing to reactivation of AR. Protein tyrosine kinases have been implicated in supporting AR activation under castrate conditions. Here we report that Lyn tyrosine kinase expression is upregulated in CRPC human specimens compared with hormone naive or normal tissue. Lyn overexpression enhanced AR transcriptional activity both in vitro and in vivo and accelerated CRPC. Reciprocally, specific targeting of Lyn resulted in a decrease of AR transcriptional activity in vitro and in vivo and prolonged time to castration. Mechanistically, we found that targeting Lyn kinase induces AR dissociation from the molecular chaperone Hsp90, leading to its ubiquitination and proteasomal degradation. This work indicates a novel mechanism of regulation of AR stability and transcriptional activity by Lyn and justifies further investigation of the Lyn tyrosine kinase as a therapeutic target for the treatment of CRPC.Oncogenesis (2014) 3, e115; doi:10.1038/oncsis.2014.30; published online 18 August 2014.
- Published
- 2014
22. Hsp27 regulates EGF/β-catenin mediated epithelial to mesenchymal transition in prostate cancer
- Author
-
Thomas, Cordonnier, Jennifer L, Bishop, Masaki, Shiota, Ka Mun, Nip, Daksh, Thaper, Sepideh, Vahid, Devon, Heroux, Martin, Gleave, and Amina, Zoubeidi
- Subjects
Male ,Epithelial-Mesenchymal Transition ,Glycogen Synthase Kinase 3 beta ,Epidermal Growth Factor ,Active Transport, Cell Nucleus ,HSP27 Heat-Shock Proteins ,Prostatic Neoplasms ,Glycogen Synthase Kinase 3 ,Matrix Metalloproteinase 9 ,Cell Movement ,Cell Line, Tumor ,Humans ,Neoplasm Invasiveness ,Snail Family Transcription Factors ,Proto-Oncogene Proteins c-akt ,Heat-Shock Proteins ,beta Catenin ,Molecular Chaperones ,Transcription Factors - Abstract
Increased expression of the molecular chaperone Hsp27 is associated with the progression of prostate cancer (PCa) to castration-resistant disease, which is lethal due to metastatic spread of the prostate tumor. Metastasis requires epithelial to mesenchymal transition (EMT), which endows cancer cells with the ability to disseminate from the primary tumor and colonize new tissue sites. A wide variety of secreted factors promote EMT, and while overexpression and constitutive activation of epidermal growth factor (EGF) signaling is associated with poor prognosis of PCa, a precise role of EGF in PCa progression to metastasis remains unclear. Here, we show that Hsp27 is required for EGF-induced cell migration, invasion and MMPs activity as well as the expression of EMT markers including Fibronectin, Vimentin and Slug with concomitant decrease of E-cadherin. Mechanistically, we found that Hsp27 is required for EGF-induced AKT and GSK3β phosphorylation and β-catenin nuclear translocation. Moreover, silencing Hsp27 decreases EGF dependent phosphorylation of β-catenin on tyrosine 142 and 654, enhances β-catenin ubiquitination and degradation, prevents β-catenin nuclear translocation and binding to the Slug promoter. These data suggest that Hsp27 is required for EGF-mediated EMT via modulation of the β-catenin/Slug signaling pathway. Together, our findings underscore the importance of Hsp27 in EGF induced EMT in PCa and highlight the use of Hsp27 knockdown as a useful strategy for patients with advanced disease.
- Published
- 2014
23. Abstract 3342: Galiellalactone derivative targets stem cell population in ENZ-resistant prostate cancer through inhibition of STAT3
- Author
-
Krisi Ketola, Micaela Janse Van Rensburg, Kate Gibson, Amina Zoubeidi, Daksh Thaper, Sepideh Vahid, Jennifer L. Bishop, Alistair Davies, and Martin Johansson
- Subjects
Cancer Research ,education.field_of_study ,biology ,Population ,CD44 ,Cancer ,medicine.disease ,Stem cell marker ,Prostate cancer ,chemistry.chemical_compound ,Oncology ,SOX2 ,chemistry ,Immunology ,Cancer research ,medicine ,biology.protein ,Enzalutamide ,Stem cell ,education - Abstract
Background: Androgen ablation remains the most effective therapy for patients with advanced disease. Unfortunately, most patients progress to castrate resistant prostate cancer (CRPC) characterized with hyper-activation of the androgen receptor (AR). Enzalutamide, a potent AR inhibitor showed efficacy by prolonging survival in CRPC patients. However, ENZ resistance (ENZR) rapidly occurs in patients and in our pre-clinical model targeting AR in ENZ resistant tumors with a 3rd generation AR inhibitor is short lived. The role of signal transducer and activator of transcription (STAT) 3 in the progression of prostate cancer is well established and has been repeatedly linked with maintenance of a stem cell phenotype across cancers. Additionally, drug resistance has been hypothesized to occur via enrichment of cancer stem-like cells (CSC), a phenotype associated with poor survival in patients. Therefore, we propose a shift of focus to target CSC phenotype using small molecule inhibitor of STAT3 called GPA500, instead of the AR axis to deal with ENZ resistance. Methods and Results: We developed a unique model of ENZ-resistance and found that cell lines derived from serially passaged ENZR tumors displayed broad genetic diversity and differential AR activity. Notably, the cell lines 42DENZR and 42FENZR are PSAlow, harbor an expanded CSC population and have STAT3 hyper-activation compared to CRPC controls measured by STAT3-luc reporter. Accordingly, Crystal Violet and MTT proliferation assays showed that 42DENZR and 42FENZR cell lines were more sensitive to the STAT3 inhibitor GPA500 compared to CRPC control. Targeting the ENZ-R cells with GPA500 reduced mRNA levels of CD133, CD44, SOX2, OCT4 and Nanog. The reduction of these CSC markers was accompanied by reduced self-renewal capacity measured by spheroid assay. Cytometry analysis revealed that treatment with GPA500 reduced α2β1+, CD44+ and CD133+ (CSC) population in the 42D and 42F cells. Conclusion: In this study, we provide pre-clinical proof that targeting the STAT3 pathway using GPA500 in ENZ resistance as well as CRPC reduces cell proliferation as well as expression of stem cell markers. Impact: Exploring mechanisms of ENZR resistance serves a critical unmet need in PCa oncology, as the number of men with ENZ resistant CRPC continues to rise. Targeting STAT3 with the small molecule inhibitor GPA500 may provide an effective method to treat ENZR patients or delay the emergence of ENZ resistance in CRPC by reducing the emergence of stem cells. Citation Format: Sepideh Vahid, Daksh Thaper, Alistair Davies, Micaela Janse Van Rensburg, Kate Gibson, Krisi Ketola, Martin Johansson, Jennifer L. Bishop, Amina Zoubeidi. Galiellalactone derivative targets stem cell population in ENZ-resistant prostate cancer through inhibition of STAT3. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3342.
- Published
- 2016
- Full Text
- View/download PDF
24. Abstract 1682: Lyn drives cancer metastasis via post-translational regulation of SNAI proteins
- Author
-
Morgan E. Roberts, Jennifer L. Bishop, Krisi Ketola, Sebastian Frees, Ka Mun Nip, Kenneth W. Harder, Daksh Thaper, Sepideh Vahid, Igor Moskalev, and Amina Zoubeidi
- Subjects
Cancer Research ,Cancer ,Biology ,medicine.disease ,Metastasis ,Oncology ,LYN ,LNCaP ,Cancer research ,medicine ,Epithelial–mesenchymal transition ,Protein kinase B ,Tyrosine kinase ,Cellular localization - Abstract
Introduction: Metastasis is the most common cause of death from cancer and occurs when malignant cells discard epithelial restraints and acquire invasive abilities, facilitating their dissemination to permissive micro-environments. This process is enhanced by tumor cell activation of Epithelial Mesenchymal Transition (EMT), a (normally embryonic) developmental program in which epithelial cells assume a mesenchymal phenotype during gastrulation and organogenesis, allowing single cell invasive movement away from the ectodermal layer. Recent evidence strongly implicates EMT induction in malignant progression and treatment resistance. For example, EMT regulatory transcription factors are required for breast cancer metastasis. Several oncogenic pathways (growth factors, Src family, MAPK, AKT) induce EMT. Lyn tyrosine kinase, a member of Src family tyrosine kinase is up-regulated in advanced prostate cancer and has been reported to correlate with aggressive breast cancer. Our objective is to determine the role of Lyn tyrosine kinase in EMT. Methods: LNCaP (Lymph Node Metastasis of Prostate Cancer), BT-549 (Triple Negative Breast Cancer) and UM-UC-13 (Bladder Cancer) cells were transfected with Lyn siRNA; EMT markers were monitored by western blot and qRT-PCR and immunofluorescence, migration by scratch assay and invasion by Boyden chamber. Sub cellular localization of proteins was examined by IF and nuclear/cyto extraction. In vivo experiments were performed in UC13-luc cells with shRNA of Lyn. Results: Here we report that Lyn expression is low in epithelial cells and is up-regulated in mesenchymal cells. Targeting Lyn using siRNA decreases EMT markers (Fibronectin, Vimentin and Zeb-1) at both mRNA and protein levels while increasing the epithelial marker (E-cadherin). Moreover, we found that Lyn siRNA decreases cell migration and invasion. Thisis decrease in mesenchymal phenotype can be attributed to the decrease in the amount of Slug and Snail, transcriptional repressors of E-Cadherin and activator of Vimentin and Fibronectin. Interestingly, we found that Lyn knockout induces a decrease of SLUG only at protein levels and not at mRNA levels. We discovered that Lyn triggers a signaling cascade through Vav-Rac-Pak1 pathway to alter sub cellular localization of the SNAI proteins leading to their proteasomal degradation. This effect results in decreased invasion and migration in vitro as well as decreased metastasis in vivo. Conclusion: Expression of Lyn kinase can be correlated to low prognosis and aggressive/metastatic phenotype. We show that knocking down Lyn by siRNA initiates a switch to a more epithelial phenotype reducing cell migration and invasion. Citation Format: Daksh Thaper, Sepideh Vahid, Ka Mun Nip, Igor Moskalev, Sebastian Frees, Morgan E. Roberts, Krisi Ketola, Kenneth W. Harder, Jennifer L. Bishop, Amina Zoubeidi. Lyn drives cancer metastasis via post-translational regulation of SNAI proteins. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1682.
- Published
- 2016
- Full Text
- View/download PDF
25. Abstract 4956: Hsp27 negatively affects Hippo tumor suppressor pathway to regulate cell survival in cancer
- Author
-
Daksh Thaper, Amina Zoubeidi, and Sepideh Vahid
- Subjects
endocrine system ,Cancer Research ,Hippo signaling pathway ,animal structures ,biology ,Cell growth ,Cancer ,medicine.disease ,Prostate cancer ,Oncology ,Hsp27 ,Cancer cell ,Immunology ,biology.protein ,Cancer research ,medicine ,TEAD1 ,Triple-negative breast cancer - Abstract
Introduction: Heat shock protein 27 (Hsp27) is a molecular chaperone highly and uniformly expressed in treatment resistant cancers like castrate resistant prostate cancer (CRPC). Hsp27 regulates activity of several oncogenic pathways and its levels correlate with aggressive tumor behaviour, drug resistance and tumor growth. Similarly, dysregulation of the Hippo tumor suppressor pathway, which restricts organ size and cell proliferation, occurs in many types of cancers. In healthy cells, activation of the Hippo pathway results in phosphorylation and cytoplasmic retention of two transcriptional co-activators YAP and TAZ, whereas in cancer YAP/TAZ are free to translocate to the nucleus and increase cell proliferation by promoting the activities of certain transcriptional factors including TEAD1. Inactivation of the Hippo pathway correlates with poor patient outcome and progression of tumors as well as an increase in migration, invasion and metastatic potential of cancer cells. Therefore it is of great importance to establish the correlation between Hsp27 and the Hippo pathway to further discover suitable targets in the treatment of metastatic malignancies. Methods: Hsp27 gain and loss of function experiments were done on 3 different cancer cell lines and the functional effects on every step of the pathway were monitored via Western blots and Immunofluorescence. Activity of YAP/TAZ after Hsp27 gain and loss of function was monitored by conducting qRT-PCR on TEAD target genes. Transcriptional activity of TEAD1 was also examined using a TEAD-dependent Luciferase reporter construct. Co-immunoprecipitation assay was conducted to analyze protein interactions in the absence/presence of Hsp27. Pathway activity in prostate cancer will be assessed by immunohistochemistry staining of core components of the pathway in patients’ tissue samples. Results: Our preliminary findings indicate that Hsp27 negatively affects the Hippo pathway. We found that targeting Hsp27 using siRNA in the PC3 (prostate cancer), A549 (lung cancer) and MDA-MB-453 (triple negative breast cancer) leads to increased cytoplasmic retention of p-YAP compared to control siRNA treated cells. Moreover, using immunofluorescence, we observed reduced nuclear translocation of the YAP/TAZ as well as sequestration of these components with cytoplasmic 14-3-3 proteins in siRNA treated PC3 cells. Furthermore inhibition of Hsp27 in prostate and lung tumour cells resulted in suppression of TEAD transcriptional activity analyzed by qRT-PCR and luciferase assay. Hsp27 over-expression experiments yielded opposite results compared to knockdown. Conclusion: Hsp27 overexpression contributes to inactivation of Hippo pathway. Targeting Hsp27 leads to inactivation of YAP and TAZ onco-proteins affecting cancer cell survival. Impact: Our data further supports the significance of targeting Hsp27 as a treatment option in cancers, especially metastatic malignancies like CRPC. Citation Format: Sepideh Vahid, Daksh Thaper, Amina Zoubeidi. Hsp27 negatively affects Hippo tumor suppressor pathway to regulate cell survival in 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 4956. doi:10.1158/1538-7445.AM2015-4956
- Published
- 2015
- Full Text
- View/download PDF
26. Abstract 4106: Lyn kinase promotes metastasis through EMT in cancers
- Author
-
Amina Zoubeidi, Jennifer L. Bishop, Daksh Thaper, Sepideh Vahid, Kirsi Ketola, and Ka Mun Nip
- Subjects
Cancer Research ,Cancer ,SRC Family Tyrosine Kinase ,Biology ,medicine.disease ,Metastasis ,Oncology ,LYN ,LNCaP ,Cancer research ,medicine ,Epithelial–mesenchymal transition ,Protein kinase B ,Tyrosine kinase - Abstract
Introduction: Metastasis is the most common cause of death from cancer and occurs when malignant cells discard epithelial restraints and acquire invasive abilities, facilitating their dissemination to permissive micro-environments. This process is enhanced by tumor cell activation of Epithelial Mesenchymal Transition (EMT), a (normally embryonic) developmental program in which epithelial cells assume a mesenchymal phenotype during gastrulation and organogenesis, allowing single cell invasive movement away from the ectodermal layer. Recent evidence strongly implicates EMT induction in malignant progression and treatment resistance. For example, EMT regulatory transcription factors are required for breast cancer metastasis. Several oncogenic pathways (growth factors, Src family, MAPK, AKT) induce EMT. Lyn tyrosine kinase, a member of Src family tyrosine kinase is up-regulated in advanced prostate cancer and has been reported to correlate with aggressive breast cancer. Our objective is to determine the role of Lyn tyrosine kinase in EMT. Methods: LNCaP, BT-549, UC-13 cells were transfected with Lyn siRNA; EMT markers were monitored by western blot and qRT-PCR and immunofluorescence, migration by scratch assay, invasion by Boyden chamber and matrix metalloproteinase (MMP) activity by Zymography. Lyn specific Kinase inhibitor Bafetinib will also be used along with over-expression experiments with Lyn Kinase Dead and Constitutively active mutants. Results: Here we report that Lyn expression is low in epithelial cells and is up-regulated in mesenchymal cells. Targeting Lyn using siRNA decreases EMT markers (Fibronectin, Vimentin) at both mRNA and protein levels while increasing the epithelial marker (E-cadherin). Moreover, we found that targeting Lyn by siRNA/shRNA and small molecule inhibitor Bafetinib decreases cell migration, invasion and the activity of MMPs. This decrease in mesenchymal phenotype can be attributed to the decrease in the amount of Slug and Snail, transcriptional repressors of E-Cadherin and activators of Vimentin. Consequently, we also found that over-expressing Lyn in both as WT or constitutively active (CA) mutant rescues expression of Slug and Snail, inducing EMT, increased cell migration and invasion while Kinase Dead (KD) mutant has no effect. Conclusion: Expression of Lyn kinase can be correlated to low prognosis and aggressive/metastatic phenotype. We show that targeting Lyn activity initiates a switch to a more epithelial phenotype reducing cell migration and invasion. Impact: The data suggests that Lyn tyrosine kinase plays a role in Epithelial Mesenchymal Transition and could be considered as a target for metastatic disease; especially in the more aggressive forms of cancer like Triple Negative Breast Cancer or Castration Resistant Prostate Cancer. This could be a realistic therapeutic option, as the Lyn small molecule inhibitor Bafetinib, is currently in clinical trials for treatment of several cancers. Note: This abstract was not presented at the meeting. Citation Format: Daksh Thaper, Sepideh Vahid, Ka Mun Nip, Kirsi Ketola, Jennifer Bishop, Amina Zoubeidi. Lyn kinase promotes metastasis through EMT in cancers. [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 4106. doi:10.1158/1538-7445.AM2015-4106
- Published
- 2015
- Full Text
- View/download PDF
27. Abstract 728: New derivative of galiellalactone inhibits the STAT3 activity and suppresses ENZ-resistant Prostate Cancer in vitro
- Author
-
Sepideh Vahid, Martin Johansson, Daksh Thaper, Amina Zoubeidi, and Jennifer L. Bishop
- Subjects
Cancer Research ,biology ,Chemistry ,Cell growth ,CD44 ,Cancer ,urologic and male genital diseases ,medicine.disease ,Androgen receptor ,Prostate cancer ,chemistry.chemical_compound ,Oncology ,Cancer stem cell ,LNCaP ,medicine ,biology.protein ,Cancer research ,Enzalutamide - Abstract
Background: The role of signal transducer and activator of transcription (STAT) 3 in the progression of Prostate Cancer (PCa), the most common male cancer in North America, is well established. Although androgen ablation remains the most effective therapy for patients with advanced disease, most progress to castrate resistant prostate cancer (CRPC). Despite the development of new potent anti-androgens, like Enzalutamide (ENZ), which prolongs survival in CRPC patients, ENZ resistance rapidly occurs. Re-activation of the androgen receptor (AR) is a hallmark of CRPC and can occur via different mechanisms, including STAT3 activation. Furthermore, STAT3 can promote CRPC independently of AR activity, however whether STAT3 promotes resistance to ENZ in CRPC remains unknown. We have found that our LNCaP xenograft derived ENZR cell lines exhibit higher activity of the STAT3 pathway compared to CRPC. Additionally, in this model further targeting the AR using novel anti-androgens is short lived or ineffective. Therefore, we propose that targeting STAT3 using the small molecule inhibitor GPA500, alone or in combination with anti-androgens may delay or treat ENZR CRPC. Methods and Results: We developed a unique model of ENZ-resistance and found that cell lines derived from serially passaged ENZR tumors displayed broad genetic diversity and differential AR activity but consistent STAT3 hyper-activation compared to CRPC controls. WB analysis showed that all ENZR cell lines had increased pSTAT3Y705 compared to CRPC. Accordingly, Crystal Violet and MTT proliferation assays showed that ENZR cell lines were more sensitive to the STAT3 inhibitor GPA500 compared to CRPC control. The reduction in growth of GPA500 treated ENZR cells was independent of PARP cleavage, whereas GPA500 induced PARP cleavage in CRPC cells. Moreover, in ENZR cell lines with high AR activity and CRPC control cells, inhibition of STAT3 reduced AR activity as shown with WB and qRT-PCR for AR downstream targets like PSA and an AR-luciferase reporter assay. In ENZR cells with low AR activity and a stem cell phenotype, GPA500 treatment reduced surface expression of α2β1, CD44 and CD133. Inhibition of STAT3 activity was confirmed by WB as well as qRT-PCR analysis of STAT3 regulated genes like c-Myc, MCL1, BCL-XL. Conclusion: In this study, we provide pre-clinical proof that targeting the STAT3 pathway using GPA500 in ENZ resistance as well as CRPC reduces cell proliferation and AR activity as well as expression of stem cell markers. Impact: Exploring mechanisms of ENZR resistance serves a critical unmet need in PCa oncology, as the number of men with ENZ resistant CRPC continues to rise. Targeting STAT3 with the small molecule inhibitor GPA500 may provide an effective method to treat ENZR patients or delay the emergence of ENZ resistance in CRPC via reduction of AR activity and/or by suppressing aggressive tumor cell phenotypes such as cancer stem cells. Citation Format: Daksh Thaper, Sepideh Vahid, Jennifer L. Bishop, Martin Johansson, Amina Zoubeidi. New derivative of galiellalactone inhibits the STAT3 activity and suppresses ENZ-resistant Prostate Cancer in vitro. [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 728. doi:10.1158/1538-7445.AM2015-728
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.