1
|
Valencia I, Nuzzo PV, Francini E, Ravera F, Fanelli GN, Bleve S, Scatena C, Marchionni L, Omar M. Gene Signature for Predicting Metastasis in Prostate Cancer Using Primary Tumor Expression Profiles. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.30.24312735. [PMID: 39252915 PMCID: PMC11383506 DOI: 10.1101/2024.08.30.24312735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Prostate cancer (PCa) is currently the most commonly diagnosed cancer and second leading cause of cancer-related death in men in the United States. The development of metastases is associated with a poor prognosis in PCa patients. Since current clinicopathological classification schemes are unable to accurately prognosticate the risk of metastasis for those diagnosed with localized PCa, there is a pressing need for precise and easily attainable biomarkers of metastatic risk in these patients. Primary tumor samples from 1239 individuals with PCa were divided into development (n=1000) and validation (n=239) cohorts. In the development cohort, we utilized a meta-analysis workflow on retrospective primary tumor gene expression profiles to identify a subset of genes predictive of metastasis. For each gene, we computed Hedges' g effect size and combined their p-values using Fisher's combined probability test. We then adjusted for multiple hypothesis testing using the Benjamini-Hochberg method. Our developed gene signature, termed Meta-Score, achieved a robust performance at predicting metastasis from primary tumor gene expression profiles, with an AUC of 0.72 in the validation cohort. In addition to its robust predictive power, Meta-Score also demonstrated a significant prognostic utility in two independent cohorts. Specifically, patients with a higher risk-score had a significantly worse metastasis-free survival and progression-free survival compared to those with lower score. Multivariate cox proportional hazards model showed that Meta-Score is significantly associated with worse survival even after adjusting for Gleason score. Our findings suggest that our primary tumor transcriptional signature, Meta-Score, could be a valuable tool to assess the risk of metastasis in PCa patients with localized disease, pending validation in large prospective studies. Author Summary Metastasis is the leading cause of death in patients diagnosed with prostate cancer (PCa), underscoring the need for reliable prediction tools to forecast the risk of metastasis at an early stage. Here, we utilize the gene expression profiles of 1,000 unique primary tumors from patients with localized PCa to develop a gene signature capable of predicting metastasis. Our signature, termed Meta-Score, comprises forty-five genes that can accurately distinguish primary tumor with high propensity for metastasis across different patient cohorts. Notably, Meta-Score maintained its robust predictive performance in an internal validation cohort of comprising primary tumor samples from 239 patients. In addition to its robust predictive performance, Meta-Score demonstrates a significant association with survival, independent of Gleason score in two independent patient cohorts, underscoring its prognostic utility. Taken together, Meta-Score is a robust risk-stratification tool that can be leveraged to identify patients at high-risk of metastasis and unfavorable survival using their primary tumor gene expression profiles.
Collapse
|
2
|
Kouroukli O, Bravou V, Giannitsas K, Tzelepi V. Tissue-Based Diagnostic Biomarkers of Aggressive Variant Prostate Cancer: A Narrative Review. Cancers (Basel) 2024; 16:805. [PMID: 38398199 PMCID: PMC10887410 DOI: 10.3390/cancers16040805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Prostate cancer (PC) is a common malignancy among elderly men, characterized by great heterogeneity in its clinical course, ranging from an indolent to a highly aggressive disease. The aggressive variant of prostate cancer (AVPC) clinically shows an atypical pattern of disease progression, similar to that of small cell PC (SCPC), and also shares the chemo-responsiveness of SCPC. The term AVPC does not describe a specific histologic subtype of PC but rather the group of tumors that, irrespective of morphology, show an aggressive clinical course, dictated by androgen receptor (AR) indifference. AR indifference represents an adaptive response to androgen deprivation therapy (ADT), driven by epithelial plasticity, an inherent ability of tumor cells to adapt to their environment by changing their phenotypic characteristics in a bi-directional way. The molecular profile of AVPC entails combined alterations in the tumor suppressor genes retinoblastoma protein 1 (RB1), tumor protein 53 (TP53), and phosphatase and tensin homolog (PTEN). The understanding of the biologic heterogeneity of castration-resistant PC (CRPC) and the need to identify the subset of patients that would potentially benefit from specific therapies necessitate the development of prognostic and predictive biomarkers. This review aims to discuss the possible pathophysiologic mechanisms of AVPC development and the potential use of emerging tissue-based biomarkers in clinical practice.
Collapse
Affiliation(s)
- Olga Kouroukli
- Department of Pathology, Evaggelismos General Hospital, 10676 Athens, Greece
| | - Vasiliki Bravou
- Department of Anatomy-Histology-Embryology, School of Medicine, University of Patras, 26504 Patras, Greece;
| | | | - Vasiliki Tzelepi
- Department of Pathology, School of Medicine, University of Patras, 26504 Patras, Greece
| |
Collapse
|
3
|
Sreekumar A, Saini S. Role of transcription factors and chromatin modifiers in driving lineage reprogramming in treatment-induced neuroendocrine prostate cancer. Front Cell Dev Biol 2023; 11:1075707. [PMID: 36711033 PMCID: PMC9879360 DOI: 10.3389/fcell.2023.1075707] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/02/2023] [Indexed: 01/14/2023] Open
Abstract
Therapy-induced neuroendocrine prostate cancer (NEPC) is a highly lethal variant of prostate cancer that is increasing in incidence with the increased use of next-generation of androgen receptor (AR) pathway inhibitors. It arises via a reversible trans-differentiation process, referred to as neuroendocrine differentiation (NED), wherein prostate cancer cells show decreased expression of AR and increased expression of neuroendocrine (NE) lineage markers including enolase 2 (ENO2), chromogranin A (CHGA) and synaptophysin (SYP). NEPC is associated with poor survival rates as these tumors are aggressive and often metastasize to soft tissues such as liver, lung and central nervous system despite low serum PSA levels relative to disease burden. It has been recognized that therapy-induced NED involves a series of genetic and epigenetic alterations that act in a highly concerted manner in orchestrating lineage switching. In the recent years, we have seen a spurt in research in this area that has implicated a host of transcription factors and epigenetic modifiers that play a role in driving this lineage switching. In this article, we review the role of important transcription factors and chromatin modifiers that are instrumental in lineage reprogramming of prostate adenocarcinomas to NEPC under the selective pressure of various AR-targeted therapies. With an increased understanding of the temporal and spatial interplay of transcription factors and chromatin modifiers and their associated gene expression programs in NEPC, better therapeutic strategies are being tested for targeting NEPC effectively.
Collapse
|
4
|
Wang Z, Wang T, Hong D, Dong B, Wang Y, Huang H, Zhang W, Lian B, Ji B, Shi H, Qu M, Gao X, Li D, Collins C, Wei G, Xu C, Lee HJ, Huang J, Li J. Single-cell transcriptional regulation and genetic evolution of neuroendocrine prostate cancer. iScience 2022; 25:104576. [PMID: 35789834 PMCID: PMC9250006 DOI: 10.1016/j.isci.2022.104576] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/24/2022] [Accepted: 06/07/2022] [Indexed: 12/30/2022] Open
Abstract
Neuroendocrine prostate cancer (NEPC) is a lethal subtype of prostate cancer, with a 10% five-year survival rate. However, little is known about its origin and the mechanisms governing its emergence. Our study characterized ADPC and NEPC in prostate tumors from 7 patients using scRNA-seq. First, we identified two NEPC gene expression signatures representing different phases of trans-differentiation. New marker genes we identified may be used for clinical diagnosis. Second, integrative analyses combining expression and subclonal architecture revealed different paths by which NEPC diverges from the original ADPC, either directly from treatment-naïve tumor cells or from specific intermediate states of treatment-resistance. Third, we inferred a hierarchical transcription factor (TF) network underlying the progression, which involves constitutive regulation by ASCL1, FOXA2, and selective regulation by NKX2-2, POU3F2, and SOX2. Together, these results defined the complex expression profiles and advanced our understanding of the genetic and transcriptomic mechanisms leading to NEPC differentiation. Single-cell RNA sequencing revealed two distinct transcriptional programs of NEPC Cell-level clonal evolution analysis extended the divergent model of ADPC to NEPC Screening of NEPC-specific transcription factors through network-based approaches
Collapse
|
5
|
Hasan MF, Ganapathy K, Sun J, Khatib A, Andl T, Soulakova JN, Coppola D, Zhang W, Chakrabarti R. LncRNA PAINT is associated with aggressive prostate cancer and dysregulation of cancer hallmark genes. Int J Cancer 2021; 149:10.1002/ijc.33569. [PMID: 33729568 PMCID: PMC9211384 DOI: 10.1002/ijc.33569] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 12/26/2022]
Abstract
Long noncoding RNAs (lncRNAs) play regulatory role in cellular processes and their aberrant expression may drive cancer progression. Here we report the function of a lncRNA PAINT (prostate cancer associated intergenic noncoding transcript) in promoting prostate cancer (PCa) progression. Upregulation of PAINT was noted in advanced stage and metastatic PCa. Inhibition of PAINT decreased cell proliferation, S-phase progression, increased expression of apoptotic markers, and improved sensitivity to docetaxel and Aurora kinase inhibitor VX-680. Inhibition of PAINT decreased cell migration and reduced expression of Slug and Vimentin. Ectopic expression of PAINT suppressed E-cadherin, increased S-phase progression and cell migration. PAINT expression in PCa cells induced larger colony formation, increased tumor growth and higher expression of mesenchymal markers. Transcriptome analysis followed by qRT-PCR validation showed differentially expressed genes involved in epithelial mesenchymal transition (EMT), apoptosis and drug resistance in PAINT-expressing cells. Our study establishes an oncogenic function of PAINT in PCa.
Collapse
Affiliation(s)
- Md Faqrul Hasan
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida
| | - Kavya Ganapathy
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida
| | - Jiao Sun
- Department of Computer Science, University of Central Florida, Orlando, Florida
| | - Ayman Khatib
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida
| | - Thomas Andl
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida
| | - Julia N. Soulakova
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida
| | - Domenico Coppola
- Moffitt Cancer Center, Tampa, Florida
- Florida Digestive Health Specialists, Bradenton, Florida
| | - Wei Zhang
- Department of Computer Science, University of Central Florida, Orlando, Florida
| | - Ratna Chakrabarti
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida
| |
Collapse
|
6
|
Kaarijärvi R, Kaljunen H, Ketola K. Molecular and Functional Links between Neurodevelopmental Processes and Treatment-Induced Neuroendocrine Plasticity in Prostate Cancer Progression. Cancers (Basel) 2021; 13:cancers13040692. [PMID: 33572108 PMCID: PMC7915380 DOI: 10.3390/cancers13040692] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Treatment-induced neuroendocrine prostate cancer (t-NEPC) is a subtype of castration-resistant prostate cancer (CRPC) which develops under prolonged androgen deprivation therapy. The mechanisms and pathways underlying the t-NEPC are still poorly understood and there are no effective treatments available. Here, we summarize the literature on the molecules and pathways contributing to neuroendocrine phenotype in prostate cancer in the context of their known cellular neurodevelopmental processes. We also discuss the role of tumor microenvironment in neuroendocrine plasticity, future directions, and therapeutic options under clinical investigation for neuroendocrine prostate cancer. Abstract Neuroendocrine plasticity and treatment-induced neuroendocrine phenotypes have recently been proposed as important resistance mechanisms underlying prostate cancer progression. Treatment-induced neuroendocrine prostate cancer (t-NEPC) is highly aggressive subtype of castration-resistant prostate cancer which develops for one fifth of patients under prolonged androgen deprivation. In recent years, understanding of molecular features and phenotypic changes in neuroendocrine plasticity has been grown. However, there are still fundamental questions to be answered in this emerging research field, for example, why and how do the prostate cancer treatment-resistant cells acquire neuron-like phenotype. The advantages of the phenotypic change and the role of tumor microenvironment in controlling cellular plasticity and in the emergence of treatment-resistant aggressive forms of prostate cancer is mostly unknown. Here, we discuss the molecular and functional links between neurodevelopmental processes and treatment-induced neuroendocrine plasticity in prostate cancer progression and treatment resistance. We provide an overview of the emergence of neurite-like cells in neuroendocrine prostate cancer cells and whether the reported t-NEPC pathways and proteins relate to neurodevelopmental processes like neurogenesis and axonogenesis during the development of treatment resistance. We also discuss emerging novel therapeutic targets modulating neuroendocrine plasticity.
Collapse
|
7
|
Vue TY, Kollipara RK, Borromeo MD, Smith T, Mashimo T, Burns DK, Bachoo RM, Johnson JE. ASCL1 regulates neurodevelopmental transcription factors and cell cycle genes in brain tumors of glioma mouse models. Glia 2020; 68:2613-2630. [PMID: 32573857 PMCID: PMC7587013 DOI: 10.1002/glia.23873] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/08/2020] [Accepted: 05/29/2020] [Indexed: 12/22/2022]
Abstract
Glioblastomas (GBMs) are incurable brain tumors with a high degree of cellular heterogeneity and genetic mutations. Transcription factors that normally regulate neural progenitors and glial development are aberrantly coexpressed in GBM, conferring cancer stem‐like properties to drive tumor progression and therapeutic resistance. However, the functional role of individual transcription factors in GBMs in vivo remains elusive. Here, we demonstrate that the basic‐helix–loop–helix transcription factor ASCL1 regulates transcriptional targets that are central to GBM development, including neural stem cell and glial transcription factors, oncogenic signaling molecules, chromatin modifying genes, and cell cycle and mitotic genes. We also show that the loss of ASCL1 significantly reduces the proliferation of GBMs induced in the brain of a genetically relevant glioma mouse model, resulting in extended survival times. RNA‐seq analysis of mouse GBM tumors reveal that the loss of ASCL1 is associated with downregulation of cell cycle genes, illustrating an important role for ASCL1 in controlling the proliferation of GBM.
Collapse
Affiliation(s)
- Tou Yia Vue
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Rahul K Kollipara
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mark D Borromeo
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Tyler Smith
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Tomoyuki Mashimo
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Dennis K Burns
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Robert M Bachoo
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jane E Johnson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
8
|
Heng BC, Gong T, Wang S, Lim LW, Wu W, Zhang C. Decellularized Matrix Derived from Neural Differentiation of Embryonic Stem Cells Enhances the Neurogenic Potential of Dental Follicle Stem Cells. J Endod 2018; 43:409-416. [PMID: 28231979 DOI: 10.1016/j.joen.2016.10.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/07/2016] [Accepted: 10/22/2016] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Dental follicle stem cells (DFSCs) possess neurogenic potential because they originate from the embryonic neural crest. This study investigated whether neural differentiation of DFSCs can be enhanced by culture on decellularized matrix substrata (NSC-DECM) derived from neurogenesis of human embryonic stem cells (hESCs). METHODS The hESCs were differentiated into neural stem cells (NSCs), and NSC-DECM was extracted from confluent monolayers of NSCs through treatment with deionized water. DFSCs seeded on NSC-DECM, Geltrex, and tissue culture polystyrene (TCPS) were subjected to neural induction during a period of 21 days. Expression of early/intermediate (Musashi1, PAX6, NSE, and βIII-tubulin) and mature/late (NGN2, NeuN, NFM, and MASH1) neural markers by DFSCs was analyzed at the 7-, 14-, and 21-day time points with quantitative real-time polymerase chain reaction. Immunocytochemistry for detection of βIII-tubulin, PAX6, and NGN2 expression by DFSCs on day 7 of neural induction was also carried out. RESULTS Quantitative RT-PCR showed that expression of PAX6, Musashi1, βIII-tubulin, NSE, NGN2, and NFM by DFSCs was enhanced on NSC-DECM versus either the Geltrex or TCPS groups. Immunocytochemistry showed that DFSCs in the NSC-DECM group displayed more intense staining for βIII-tubulin, PAX6, and NGN2 expression, together with more neurite outgrowths and elongated morphology, as compared with either Geltrex or TCPS. CONCLUSIONS DECM derived from neurogenesis of hESCs can enhance the neurogenic potential of DFSCs.
Collapse
Affiliation(s)
- Boon Chin Heng
- Endodontology, Faculty of Dentistry, University of Hong Kong, Pokfulam, Hong Kong, China; Department of Biological Sciences, Sunway University, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | - Ting Gong
- Endodontology, Faculty of Dentistry, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Shuai Wang
- ENT Institute of Shenzhen, Shenzhen Longgang ENT Hospital, Shenzhen, China
| | - Lee Wei Lim
- Department of Biological Sciences, Sunway University, Bandar Sunway, Selangor Darul Ehsan, Malaysia; School of Biomedical Sciences, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Wutian Wu
- School of Biomedical Sciences, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Chengfei Zhang
- Endodontology, Faculty of Dentistry, University of Hong Kong, Pokfulam, Hong Kong, China; HKU Shenzhen Institute of Research and Innovation, Hong Kong, China.
| |
Collapse
|
9
|
Nam SM, Kim YN, Kim JW, Kyeong DS, Lee SH, Son Y, Shin JH, Kim J, Yi SS, Yoon YS, Seong JK. Hairy and Enhancer of Split 6 (Hes6) Deficiency in Mouse Impairs Neuroblast Differentiation in Dentate Gyrus Without Affecting Cell Proliferation and Integration into Mature Neurons. Cell Mol Neurobiol 2016; 36:57-67. [PMID: 26105991 DOI: 10.1007/s10571-015-0220-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 06/01/2015] [Indexed: 11/29/2022]
Abstract
Hes6 is a member of the hairy-enhancer of split homolog (Hes) family of transcription factors and interacts with other Hes family genes. During development, Hes genes are expressed in neural stem cells and progenitor cells. However, the role of Hes6 in adult hippocampal neurogenesis remains unclear. We therefore investigated the effects of Hes6 on adult hippocampal neurogenesis, by comparing Hes6 knockout and wild-type mice. To this end, we immunostained for markers of neural stem cells and progenitor cells (nestin), proliferating cells (Ki67), post-mitotic neuroblasts and immature neurons (doublecortin, DCX), mature neuronal cells (NeuN), and astrocyte (S100β). We also injected 5-bromo-2'-deoxyuridine (BrdU) to trace the fate of mitotic cells. Nestin- and Ki67-positive proliferating cells did now show any significant differences between wild and knockout groups. Hes6 knockout negatively affects neuroblast differentiation based on DCX immunohistochemistry. On the contrary, the ratio of the BrdU and NeuN double-positive cells did not show any significance, even though it was slightly higher in the knockout group. These results suggest that Hes6 is involved in the regulation of neuroblast differentiation during adult neurogenesis, but does not influence integration into mature neurons.
Collapse
Affiliation(s)
- Sung Min Nam
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Seoul National University, Seoul, 151-742, South Korea
- BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742, South Korea
- KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul, 151-742, South Korea
| | - Yo Na Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Seoul National University, Seoul, 151-742, South Korea
- BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742, South Korea
- KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul, 151-742, South Korea
| | - Jong Whi Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Seoul National University, Seoul, 151-742, South Korea
- BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742, South Korea
- KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul, 151-742, South Korea
| | - Dong Soo Kyeong
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Seoul National University, Seoul, 151-742, South Korea
- BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742, South Korea
- KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul, 151-742, South Korea
| | - Seo Hyun Lee
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Seoul National University, Seoul, 151-742, South Korea
- BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742, South Korea
- KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul, 151-742, South Korea
| | - Yeri Son
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Seoul National University, Seoul, 151-742, South Korea
- BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742, South Korea
- KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul, 151-742, South Korea
| | - Jae Hoon Shin
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Seoul National University, Seoul, 151-742, South Korea
- BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742, South Korea
- KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul, 151-742, South Korea
| | - Jaesang Kim
- Department of Life Sciences and Ewha Research Center for Systems Biology, Ewha Womans University, Seoul, 120-750, South Korea
| | - Sun Shin Yi
- Department of Biomedical Laboratory Science, College of Biomedical Sciences, Soonchunhyang University, Asan, 336-745, South Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Seoul National University, Seoul, 151-742, South Korea.
- BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742, South Korea.
- KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul, 151-742, South Korea.
| | - Je Kyung Seong
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Seoul National University, Seoul, 151-742, South Korea.
- BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742, South Korea.
- KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul, 151-742, South Korea.
- Interdisciplinary Program for Bioinformatics, Program for Cancer Biology and BIO-MAX Institute, Seoul National University, Seoul, 151-742, South Korea.
| |
Collapse
|
10
|
|
11
|
Ramos-Montoya A, Lamb AD, Russell R, Carroll T, Jurmeister S, Galeano-Dalmau N, Massie CE, Boren J, Bon H, Theodorou V, Vias M, Shaw GL, Sharma NL, Ross-Adams H, Scott HE, Vowler SL, Howat WJ, Warren AY, Wooster RF, Mills IG, Neal DE. HES6 drives a critical AR transcriptional programme to induce castration-resistant prostate cancer through activation of an E2F1-mediated cell cycle network. EMBO Mol Med 2014; 6:651-61. [PMID: 24737870 PMCID: PMC4023887 DOI: 10.1002/emmm.201303581] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Castrate-resistant prostate cancer (CRPC) is poorly characterized and heterogeneous and while the androgen receptor (AR) is of singular importance, other factors such as c-Myc and the E2F family also play a role in later stage disease. HES6 is a transcription co-factor associated with stem cell characteristics in neural tissue. Here we show that HES6 is up-regulated in aggressive human prostate cancer and drives castration-resistant tumour growth in the absence of ligand binding by enhancing the transcriptional activity of the AR, which is preferentially directed to a regulatory network enriched for transcription factors such as E2F1. In the clinical setting, we have uncovered a HES6-associated signature that predicts poor outcome in prostate cancer, which can be pharmacologically targeted by inhibition of PLK1 with restoration of sensitivity to castration. We have therefore shown for the first time the critical role of HES6 in the development of CRPC and identified its potential in patient-specific therapeutic strategies.
Collapse
Affiliation(s)
- Antonio Ramos-Montoya
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Alastair D Lamb
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK,Department of Urology, Addenbrooke's HospitalCambridge, UK,*Corresponding author. Tel: +44 1223 331940; Fax: +44 1223 769007; E-mail:
| | - Roslin Russell
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Thomas Carroll
- Bioinformatics Core Facility, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Sarah Jurmeister
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Nuria Galeano-Dalmau
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Charlie E Massie
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Joan Boren
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Helene Bon
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Vasiliki Theodorou
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Maria Vias
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Greg L Shaw
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK,Department of Urology, Addenbrooke's HospitalCambridge, UK
| | - Naomi L Sharma
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK,Department of Urology, Addenbrooke's HospitalCambridge, UK
| | - Helen Ross-Adams
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Helen E Scott
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Sarah L Vowler
- Bioinformatics Core Facility, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - William J Howat
- Histopathology/ISH Core Facility, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Anne Y Warren
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK,Department of Pathology, Addenbrooke's HospitalCambridge, UK
| | | | - Ian G Mills
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK,Prostate Cancer Research Group, Nordic EMBL Partnership, Centre for Molecular Medicine Norway (NCMM), University of OsloOslo, Norway,Departments of Cancer Prevention and Urology, Institute of Cancer Research and Oslo University HospitalsOslo, Norway
| | - David E Neal
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK,Department of Urology, Addenbrooke's HospitalCambridge, UK,Department of Oncology, University of CambridgeCambridge, UK,**Corresponding author. Tel: +44 1223 331940; Fax: +44 1223 769007; E-mail:
| |
Collapse
|
12
|
Baxendale IR, Cheung S, Kitching MO, Ley SV, Shearman JW. The synthesis of neurotensin antagonist SR 48692 for prostate cancer research. Bioorg Med Chem 2013; 21:4378-87. [PMID: 23721919 DOI: 10.1016/j.bmc.2013.04.075] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Revised: 04/18/2013] [Accepted: 04/20/2013] [Indexed: 01/03/2023]
Abstract
An improved synthesis of the molecule SR 48692 is presented and its use as a neurotensin antagonist biological probe for use in cancer research is described. The preparation includes an number of enhanced chemical conversions and strategies to overcome some of the limiting synthetic transformations in the original chemical route.
Collapse
Affiliation(s)
- I R Baxendale
- Department of Chemistry, Durham University, South Road, Durham DH1 3LE, United Kingdom.
| | | | | | | | | |
Collapse
|
13
|
A nine-gene signature predicting clinical outcome in cutaneous melanoma. J Cancer Res Clin Oncol 2012; 139:249-58. [DOI: 10.1007/s00432-012-1322-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 09/17/2012] [Indexed: 12/16/2022]
|
14
|
Wickramasinghe CM, Domaschenz R, Amagase Y, Williamson D, Missiaglia E, Shipley J, Murai K, Jones PH. HES6 enhances the motility of alveolar rhabdomyosarcoma cells. Exp Cell Res 2012; 319:103-12. [PMID: 22982728 DOI: 10.1016/j.yexcr.2012.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 08/24/2012] [Accepted: 08/26/2012] [Indexed: 01/12/2023]
Abstract
HES6, a member of the hairy-enhancer-of-split family of transcription factors, plays multiple roles in myogenesis. It is a direct target of the myogenic transcription factor MyoD and has been shown to regulate the formation of the myotome in development, myoblast cell cycle exit and the organization of the actin cytoskeleton during terminal differentiation. Here we investigate the expression and function of HES6 in rhabdomyosarcoma, a soft tissue tumor which expresses myogenic genes but fails to differentiate into muscle. We show that HES6 is expressed at high levels in the subset of alveolar rhabdomyosarcomas expressing PAX/FOXO1 fusion genes (ARMSp). Knockdown of HES6 mRNA in the ARMSp cell line RH30 reduces proliferation and cell motility. This phenotype is rescued by expression of mouse Hes6 which is insensitive to HES6 siRNA. Furthermore, expression microarray analysis indicates that the HES6 knockdown is associated with a decrease in the levels of Transgelin, (TAGLN), a regulator of the actin cytoskeleton. Knockdown of TAGLN decreases cell motility, whilst TAGLN overexpression rescues the motility defect resulting from HES6 knockdown. These findings indicate HES6 contributes to the pathogenesis of ARMSp by enhancing both proliferation and cell motility.
Collapse
|
15
|
Righi L, Rapa I, Votta A, Papotti M, Sapino A. Human achaete-scute homolog-1 expression in neuroendocrine breast carcinoma. Virchows Arch 2012; 460:415-21. [PMID: 22422124 DOI: 10.1007/s00428-012-1223-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 02/27/2012] [Accepted: 03/05/2012] [Indexed: 12/16/2022]
Abstract
Neuroendocrine (NE) breast carcinoma is defined by morphological features similar to those of NE tumors of other organs and NE marker expression in at least 50 % of neoplastic cells. However, a NE morphology may be observed even in breast carcinomas lacking NE markers. Human achaete-scute homolog-1 (hASH-1) is a transcription factor that plays a key role in the regulation of mammalian neural and NE cell development and has been identified in several human NE tumors. The aim of this study was to investigate hASH-1 expression in human breast cancers. hASH-1 expression was evaluated in 482 consecutive non-NE invasive breast carcinomas, in a series of 84 breast cancers with >50 % NE marker expression (high NE differentiation) and 21 carcinomas with NE histology but negative or focally (<50 %) positive for NE markers (low NE differentiation). hASH-1 protein was evaluated by a specific monoclonal antibody using immunohistochemistry and gene expression by real-time polymerase chain reaction. None of the non-NE invasive breast carcinomas expressed hASH-1 at any levels. hASH-1 was expressed in tumor cell nuclei of 63 and 38 % of cases with high and low NE differentiation, respectively. Strong correlation with protein and gene expression levels was observed (p < 0.0001). hASH-1 expression was correlated to a low mitotic count (p = 0.02) and a low Ki67 proliferative index (p = 0.0062). hASH-1 expression occurs in breast cancers with NE differentiation regardless of the extent of the NE cell population, and it is restricted to a subset of tumor cells having a low proliferative potential.
Collapse
Affiliation(s)
- Luisella Righi
- Department of Clinical and Biological Sciences, University of Turin at San Luigi Hospital, Orbassano, Turin, Italy.
| | | | | | | | | |
Collapse
|
16
|
Haapa-Paananen S, Kiviluoto S, Waltari M, Puputti M, Mpindi JP, Kohonen P, Tynninen O, Haapasalo H, Joensuu H, Perälä M, Kallioniemi O. HES6 gene is selectively overexpressed in glioma and represents an important transcriptional regulator of glioma proliferation. Oncogene 2012; 31:1299-310. [PMID: 21785461 DOI: 10.1038/onc.2011.316] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Revised: 06/14/2011] [Accepted: 06/21/2011] [Indexed: 12/15/2022]
Abstract
Malignant glioma is the most common brain tumor with 16,000 new cases diagnosed annually in the United States. We performed a systematic large-scale transcriptomics data mining study of 9783 tissue samples from the GeneSapiens database to systematically identify genes that are most glioma-specific. We searched for genes that were highly expressed in 322 glioblastoma multiforme tissue samples and 66 anaplastic astrocytomas as compared with 425 samples from histologically normal central nervous system. Transcription cofactor HES6 (hairy and enhancer of split 6) emerged as the most glioma-specific gene. Immunostaining of a tissue microarray showed HES6 expression in 335 (98.8%) out of the 339 glioma samples. HES6 was expressed in endothelial cells of the normal brain and glioma tissue. Recurrent grade 2 astrocytomas and grade 2 or 3 oligodendrogliomas showed higher levels of HES6 immunoreactivity than the corresponding primary tumors. High HES6 mRNA expression correlated with the proneural subtype that generally has a favorable outcome but is prone to recur. Functional studies suggested an important role for HES6 in supporting survival of glioma cells, as evidenced by reduction of cancer cell proliferation and migration after HES6 silencing. The biological role and consequences of HES6 silencing and overexpression was explored with genome-wide analyses, which implicated a role for HES6 in p53, c-myc and nuclear factor-κB transcriptional networks. We conclude that HES6 is important for glioma cell proliferation and migration, and may have a role in angiogenesis.
Collapse
Affiliation(s)
- S Haapa-Paananen
- Department of Medical Biotechnology, VTT Technical Research Centre of Finland and Centre for Biotechnology, University of Turku, Turku, Finland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Tagne JB, Gupta S, Gower AC, Shen SS, Varma S, Lakshminarayanan M, Cao Y, Spira A, Volkert TL, Ramirez MI. Genome-wide analyses of Nkx2-1 binding to transcriptional target genes uncover novel regulatory patterns conserved in lung development and tumors. PLoS One 2012; 7:e29907. [PMID: 22242187 PMCID: PMC3252372 DOI: 10.1371/journal.pone.0029907] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Accepted: 12/07/2011] [Indexed: 01/10/2023] Open
Abstract
The homeodomain transcription factor Nkx2-1 is essential for normal lung development and homeostasis. In lung tumors, it is considered a lineage survival oncogene and prognostic factor depending on its expression levels. The target genes directly bound by Nkx2-1, that could be the primary effectors of its functions in the different cellular contexts where it is expressed, are mostly unknown. In embryonic day 11.5 (E11.5) mouse lung, epithelial cells expressing Nkx2-1 are predominantly expanding, and in E19.5 prenatal lungs, Nkx2-1-expressing cells are predominantly differentiating in preparation for birth. To evaluate Nkx2-1 regulated networks in these two cell contexts, we analyzed genome-wide binding of Nkx2-1 to DNA regulatory regions by chromatin immunoprecipitation followed by tiling array analysis, and intersected these data to expression data sets. We further determined expression patterns of Nkx2-1 developmental target genes in human lung tumors and correlated their expression levels to that of endogenous NKX2-1. In these studies we uncovered differential Nkx2-1 regulated networks in early and late lung development, and a direct function of Nkx2-1 in regulation of the cell cycle by controlling the expression of proliferation-related genes. New targets, validated in Nkx2-1 shRNA transduced cell lines, include E2f3, Cyclin B1, Cyclin B2, and c-Met. Expression levels of Nkx2-1 direct target genes identified in mouse development significantly correlate or anti-correlate to the levels of endogenous NKX2-1 in a dosage-dependent manner in multiple human lung tumor expression data sets, supporting alternative roles for Nkx2-1 as a transcriptional activator or repressor, and direct regulator of cell cycle progression in development and tumors.
Collapse
Affiliation(s)
- Jean-Bosco Tagne
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Sumeet Gupta
- Center for Microarray Technology, Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
| | - Adam C. Gower
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Steven S. Shen
- Clinical and Translational Science Institute (CTSI), Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Saaket Varma
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | | | - Yuxia Cao
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Avrum Spira
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
- Clinical and Translational Science Institute (CTSI), Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Thomas L. Volkert
- Center for Microarray Technology, Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
| | - Maria I. Ramirez
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
18
|
Castro DS, Martynoga B, Parras C, Ramesh V, Pacary E, Johnston C, Drechsel D, Lebel-Potter M, Garcia LG, Hunt C, Dolle D, Bithell A, Ettwiller L, Buckley N, Guillemot F. A novel function of the proneural factor Ascl1 in progenitor proliferation identified by genome-wide characterization of its targets. Genes Dev 2011; 25:930-45. [PMID: 21536733 DOI: 10.1101/gad.627811] [Citation(s) in RCA: 311] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Proneural genes such as Ascl1 are known to promote cell cycle exit and neuronal differentiation when expressed in neural progenitor cells. The mechanisms by which proneural genes activate neurogenesis--and, in particular, the genes that they regulate--however, are mostly unknown. We performed a genome-wide characterization of the transcriptional targets of Ascl1 in the embryonic brain and in neural stem cell cultures by location analysis and expression profiling of embryos overexpressing or mutant for Ascl1. The wide range of molecular and cellular functions represented among these targets suggests that Ascl1 directly controls the specification of neural progenitors as well as the later steps of neuronal differentiation and neurite outgrowth. Surprisingly, Ascl1 also regulates the expression of a large number of genes involved in cell cycle progression, including canonical cell cycle regulators and oncogenic transcription factors. Mutational analysis in the embryonic brain and manipulation of Ascl1 activity in neural stem cell cultures revealed that Ascl1 is indeed required for normal proliferation of neural progenitors. This study identified a novel and unexpected activity of the proneural gene Ascl1, and revealed a direct molecular link between the phase of expansion of neural progenitors and the subsequent phases of cell cycle exit and neuronal differentiation.
Collapse
Affiliation(s)
- Diogo S Castro
- Medical Research Council National Institute for Medical Research, Division of Molecular Neurobiology, The Ridgeway, Mill Hill, London NW7 1AA, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Aparicio A, Tzelepi V, Araujo JC, Guo CC, Liang S, Troncoso P, Logothetis CJ, Navone NM, Maity SN. Neuroendocrine prostate cancer xenografts with large-cell and small-cell features derived from a single patient's tumor: morphological, immunohistochemical, and gene expression profiles. Prostate 2011; 71:846-56. [PMID: 21456067 PMCID: PMC3883511 DOI: 10.1002/pros.21301] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 09/26/2010] [Indexed: 01/24/2023]
Abstract
BACKGROUND Small-cell carcinoma (SCC) of the prostate is an AR-negative variant of prostate cancer found at progression in 10-20% of castrate-resistant disease. Its finding predicts a distinct clinical course and a poor prognosis. Large-cell neuroendocrine carcinoma (LCNEC) is a much rarer variant that behaves similarly to SCC. The biological mechanisms that drive these disease variants are poorly understood. METHODS Eight tumor fragments from the salvage pelvic exenteration specimen of a patient with castrate-resistant prostate carcinoma were subcutaneously implanted into 6- to 8-week-old male CB17 SCID mice. Serial tissue sections and tissue microarrays of the resulting MDA PCa 144 xenograft lines were used for histopathologic and immunohistochemical characterization of the xenografts and their tissue of origin. RNA from two representative xenograft sublines was used for gene-expression profiling. RESULTS All eight fragments formed tumors: four of the MDA PCa 144 xenograft sublines had morphologic characteristics of SCC and four, of LCNEC. All retained high fidelity to their parent tumor tissue, which remained stable through serial passages. Morphological transitions in the specimen of origin suggested LCNEC represents an intermediate step between adenocarcinoma and SCC. Over 2,500 genes were differentially expressed between the SCC (MDA PCa 144-13) and the LCNEC (MDA PCa 144-4) sublines and enriched in "Nervous System Development" Gene Ontology subtree. CONCLUSION The eight xenograft models described represent the spectrum of neuroendocrine carcinomas in prostate cancer and will be valuable preclinical tools to study the pathogenesis of and therapy targets for this increasingly recognized subset of lethal prostate cancer.
Collapse
MESH Headings
- Aged
- Animals
- Antineoplastic Agents/therapeutic use
- Carcinoma, Large Cell/drug therapy
- Carcinoma, Large Cell/pathology
- Carcinoma, Large Cell/radiotherapy
- Carcinoma, Neuroendocrine/drug therapy
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Neuroendocrine/radiotherapy
- Carcinoma, Small Cell/drug therapy
- Carcinoma, Small Cell/pathology
- Carcinoma, Small Cell/radiotherapy
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Mice
- Mice, SCID
- Prostate-Specific Antigen
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms/radiotherapy
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Ana Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
- Correspondence to: Department of Genitourinary Medical Oncology, Unit 1374, 1515 Holcombe Boulevard, Houston, TX 77030-4009. Tel: 713-563-6969; Fax: 713-745-1625;
| | - Vasiliki Tzelepi
- Department of Genitourinary Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - John C. Araujo
- Department of Genitourinary Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Charles C. Guo
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Shoudan Liang
- Department of Bioinformatics and Computational Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Patricia Troncoso
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Christopher J. Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Nora M. Navone
- Department of Genitourinary Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Sankar N. Maity
- Department of Genitourinary Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
20
|
Cindolo L, Cantile M, Franco R, Chiodini P, Schiavo G, Forte I, Zlobec I, Salzano L, Botti G, Gidaro S, Terracciano L, Cillo C. Parallel determination of NeuroD1, Chromogranin-A, KI67 and androgen receptor expression in surgically treated prostate cancers. Int Braz J Urol 2011; 37:57-66. [DOI: 10.1590/s1677-55382011000100008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2010] [Indexed: 12/23/2022] Open
Affiliation(s)
| | | | - R. Franco
- G. Pascale National Cancer Institute, Italy
| | | | | | - I. Forte
- G. Pascale National Cancer Institute, Italy
| | | | | | - G. Botti
- G. Pascale National Cancer Institute, Italy
| | | | | | - C. Cillo
- Federico II University, Italy; University of Basel, Switzerland
| |
Collapse
|
21
|
Qi J, Nakayama K, Cardiff RD, Borowsky AD, Kaul K, Williams R, Krajewski S, Mercola D, Carpenter PM, Bowtell D, Ronai ZA. Siah2-dependent concerted activity of HIF and FoxA2 regulates formation of neuroendocrine phenotype and neuroendocrine prostate tumors. Cancer Cell 2010; 18:23-38. [PMID: 20609350 PMCID: PMC2919332 DOI: 10.1016/j.ccr.2010.05.024] [Citation(s) in RCA: 188] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Revised: 03/25/2010] [Accepted: 05/14/2010] [Indexed: 11/24/2022]
Abstract
Neuroendocrine (NE) phenotype, seen in >30% of prostate adenocarcinomas (PCa), and NE prostate tumors are implicated in aggressive prostate cancer. Formation of NE prostate tumors in the TRAMP mouse model was suppressed in mice lacking the ubiquitin ligase Siah2, which regulates HIF-1alpha availability. Cooperation between HIF-1alpha and FoxA2, a transcription factor expressed in NE tissue, promotes recruitment of p300 to transactivate select HIF-regulated genes, Hes6, Sox9, and Jmjd1a. These HIF-regulated genes are highly expressed in metastatic PCa and required for hypoxia-mediated NE phenotype, metastasis in PCa, and the formation of NE tumors. Tissue-specific expression of FoxA2 combined with Siah2-dependent HIF-1alpha availability enables a transcriptional program required for NE prostate tumor development and NE phenotype in PCa.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Animals
- Cell Line, Tumor
- Female
- Gene Expression Regulation, Neoplastic
- Hepatocyte Nuclear Factor 3-beta/genetics
- Hepatocyte Nuclear Factor 3-beta/metabolism
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Liver Neoplasms, Experimental/genetics
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/secondary
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Lymphatic Metastasis
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Neuroendocrine Tumors/genetics
- Neuroendocrine Tumors/metabolism
- Neuroendocrine Tumors/pathology
- Neurosecretory Systems/metabolism
- Neurosecretory Systems/pathology
- Phenotype
- Prostate/metabolism
- Prostate/pathology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Signal Transduction
- Transcriptional Activation
- Ubiquitin-Protein Ligases/physiology
Collapse
Affiliation(s)
- Jianfei Qi
- Signal Transduction Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Wnt-11 promotes neuroendocrine-like differentiation, survival and migration of prostate cancer cells. Mol Cancer 2010; 9:55. [PMID: 20219091 PMCID: PMC2846888 DOI: 10.1186/1476-4598-9-55] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Accepted: 03/10/2010] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Wnt-11 is a secreted protein that modulates cell growth, differentiation and morphogenesis during development. We previously reported that Wnt-11 expression is elevated in hormone-independent prostate cancer and that the progression of prostate cancer from androgen-dependent to androgen-independent proliferation correlates with a loss of mutual inhibition between Wnt-11- and androgen receptor-dependent signals. However, the prevalence of increased expression of Wnt-11 in patient tumours and the functions of Wnt-11 in prostate cancer cells were not known. RESULTS Wnt-11 protein levels in prostate tumours were determined by immunohistochemical analysis of prostate tumour tissue arrays. Wnt-11 protein was elevated in 77/117 of tumours when compared with 27 benign prostatic hypertrophy specimens and was present in 4/4 bone metastases. In addition, there was a positive correlation between Wnt-11 expression and PSA levels above 10 ng/ml. Androgen-depleted LNCaP prostate cancer cells form neurites and express genes associated with neuroendocrine-like differentiation (NED), a feature of prostate tumours that have a poor prognosis. Since androgen-depletion increases expression of Wnt-11, we examined the role of Wnt-11 in NED. Ectopic expression of Wnt-11 induced expression of NSE and ASCL1, which are markers of NED, and this was prevented by inhibitors of cyclic AMP-dependent protein kinase, consistent with the known role of this kinase in NED. In contrast, Wnt-11 did not induce NSE expression in RWPE-1 cells, which are derived from benign prostate, suggesting that the role of Wnt-11 in NED is specific to prostate cancer. In addition, silencing of Wnt-11 expression in androgen-depleted LNCaP cells prevented NED and resulted in apoptosis. Silencing of Wnt-11 gene expression in androgen-independent PC3 cells also reduced expression of NSE and increased apoptosis. Finally, silencing of Wnt-11 reduced PC3 cell migration and ectopic expression of Wnt-11 promoted LNCaP cell invasion. CONCLUSIONS These observations suggest that the increased level of Wnt-11 found in prostate cancer contributes to tumour progression by promoting NED, tumour cell survival and cell migration/invasion, and may provide an opportunity for novel therapy in prostate cancer.
Collapse
|
23
|
Wang J, Kim J, Roh M, Franco OE, Hayward SW, Wills ML, Abdulkadir SA. Pim1 kinase synergizes with c-MYC to induce advanced prostate carcinoma. Oncogene 2010; 29:2477-87. [PMID: 20140016 PMCID: PMC2861731 DOI: 10.1038/onc.2010.10] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The oncogenic PIM1 kinase has been implicated as a cofactor for c-MYC in prostate carcinogenesis. In this study, we show that in human prostate tumors, coexpression of c-MYC and PIM1 is associated with higher Gleason grades. Using a tissue recombination model coupled with lentiviral-mediated gene transfer we find that Pim1 is weakly oncogenic in naive adult mouse prostatic epithelium. However, it cooperates dramatically with c-MYC to induce prostate cancer within 6-weeks. Importantly, c-MYC/Pim1 synergy is critically dependent on Pim1 kinase activity. c-MYC/Pim1 tumors showed increased levels of the active serine-62 (S62) phosphorylated form of c-MYC. Grafts expressing a phosphomimetic c-MYCS62D mutant had higher rates of proliferation than grafts expressing wild type c-MYC but did not form tumors like c-MYC/Pim1 grafts, indicating that Pim1 cooperativity with c-MYC in vivo involves additional mechanisms other than enhancement of c-MYC activity by S62 phosphorylation. c-MYC/Pim1-induced prostate carcinomas show evidence of neuroendocrine (NE) differentiation. Additional studies, including the identification of tumor cells coexpressing androgen receptor and NE cell markers synaptophysin and Ascl1 suggested that NE tumors arose from adenocarcinoma cells through transdifferentiation. These results directly show functional cooperativity between c-MYC and PIM1 in prostate tumorigenesis in vivo and support efforts for targeting PIM1 in prostate cancer.
Collapse
Affiliation(s)
- J Wang
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Hartman J, Lam EWF, Gustafsson JA, Ström A. Hes-6, an inhibitor of Hes-1, is regulated by 17beta-estradiol and promotes breast cancer cell proliferation. Breast Cancer Res 2009; 11:R79. [PMID: 19891787 PMCID: PMC2815541 DOI: 10.1186/bcr2446] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 10/03/2009] [Accepted: 11/05/2009] [Indexed: 01/30/2023] Open
Abstract
Introduction Hes-6 is a member of the basic helix-loop-helix (bHLH) family of transcription factors, and its overexpression has been reported in metastatic cancers of different origins. Hes-6 has been described as an inhibitor of Hes-1 during neuronal development, although its function in cancer is not known. In this study, we investigated the function of Hes-6 in breast cancer and tested the hypothesis that Hes-6 enhances breast cancer cell proliferation and is regulated by estrogen. Methods To investigate the function of Hes-6, T47D cells stably expressing Hes-6 were generated by lentiviral transduction, and conversely, siRNA also was used to knock down Hes-6 expression in breast cancer cells. The Hes-6-expressing T47D cells were transplanted into immunodeficient mice to study effects on tumor growth. Results We found that Hes-6 expression was significantly higher in the high-grade, estrogen receptor (ER)α-negative SKBR3 and MDA-MB-231 cells compared with the ERα-positive, non-metastasizing T47D and MCF-7 breast carcinoma cells. Moreover, the level of Hes-6 mRNA was 28 times higher in breast cancer samples compared with normal breast samples. In Hes-6-expressing T47D cells, Hes-6 ectopic expression was shown to stimulate cell proliferation in vitro as well as breast tumor growth in xenografts. Moreover, expression of Hes-6 resulted in induction of E2F-1, a crucial target gene for the transcriptional repressor Hes-1. Consistently, silencing of Hes-6 by siRNA resulted in downregulation of E2F-1 expression, whereas estrogen treatment caused induction of Hes-6 and downstream targets hASH-1 and E2F-1 in MCF-7 cells. Conclusions Together, the data suggest that Hes-6 is a potential oncogene overexpressed in breast cancer, with a tumor-promoting and proliferative function. Furthermore, Hes-6 is a novel estrogen-regulated gene in breast cancer cells. An understanding of the role and regulation of Hes-6 could provide insights into estrogen signaling and endocrine resistance in breast cancer and, hence, could be important for the development of novel anticancer drugs.
Collapse
Affiliation(s)
- Johan Hartman
- Department of Biosciences and Nutrition, Karolinska Institutet, Nobels väg 5, Solna Alfred Nobels Allé 8, 141 57 Huddinge, Sweden.
| | | | | | | |
Collapse
|
25
|
Myers RM, Shearman JW, Kitching MO, Ramos-Montoya A, Neal DE, Ley SV. Cancer, chemistry, and the cell: molecules that interact with the neurotensin receptors. ACS Chem Biol 2009; 4:503-25. [PMID: 19462983 DOI: 10.1021/cb900038e] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The literature covering neurotensin (NT) and its signalling pathways, receptors, and biological profile is complicated by the fact that the discovery of three NT receptor subtypes has come to light only in recent years. Moreover, a lot of this literature explores NT in the context of the central nervous system and behavioral studies. However, there is now good evidence that the up-regulation of NT is intimately involved in cancer development and progression. This Review aims to summarize the isolation, cloning, localization, and binding properties of the accepted receptor subtypes (NTR1, NTR2, and NTR3) and the molecules known to bind at these receptors. The growing role these targets are playing in cancer research is also discussed. We hope this Review will provide a useful overview and a one-stop resource for new researchers engaged in this field at the chemistry-biology interface.
Collapse
Affiliation(s)
- Rebecca M. Myers
- Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - James W. Shearman
- Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Matthew O. Kitching
- Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Antonio Ramos-Montoya
- CRUK-Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, United Kingdom
| | - David E. Neal
- CRUK-Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, United Kingdom
| | - Steven V. Ley
- Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
26
|
Vias M, Ramos-Montoya A, Mills IG. Terminal and progenitor lineage-survival oncogenes as cancer markers. Trends Mol Med 2008; 14:486-94. [PMID: 18929510 DOI: 10.1016/j.molmed.2008.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Revised: 09/01/2008] [Accepted: 09/01/2008] [Indexed: 12/31/2022]
Abstract
Tumour classification has traditionally focused on differentiation and cellular morphology, and latterly on the application of genomic approaches. By combining chromatin immunoprecipitation with expression array, it has been possible to identify direct gene targets for transcription factors for nuclear hormone receptors. At the same time, there have been great strides in deriving stem and progenitor cells from tissues. It is therefore timely to propose that pairing the isolation of these cell subpopulations from tissues and tumours with these genomics approaches will reveal conserved gene targets for transcription factors. By focusing on transcription factors (lineage-survival oncogenes) with roles in both organogenesis and tumourigenesis at multiple organ sites, we suggest that this comparative genomics approach will enable developmental biology to be used more fully in relation to understanding tumour progression and will reveal new cancer markers. We focus here on neurogenesis and neuroendocrine differentiation in tumours.
Collapse
Affiliation(s)
- Maria Vias
- Uro-Oncology Research Group, Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | | | | |
Collapse
|