Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Endocr Relat Cancer ; 28(8): T11-T18, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34128829

ABSTRACT

The first case of prostate cancer was identified by histological examination by Adams, a surgeon at The London Hospital, in 1853. In his report, Adams noted that the condition was 'a very rare disease'. Now, over 150 years later, with increased life expectancy and screening, prostate cancer has become one of the most common cancers in men. In the United States alone, nearly 200,000 men are diagnosed with prostate cancer annually and about 33,000 succumb to their disease. Fifty years ago, men were typically diagnosed with prostate cancer in their seventies with disease that had metastasized to the bone and/or soft tissue. Diagnosis at such an advanced stage was a death sentence, with patients dying within 2 years. The pioneering work of Charles Huggins in the 1940s found that metastatic prostate cancer responds to androgen deprivation therapy (ADT), ushering in the rational use of hormone therapies that have irrevocably changed the course of prostate cancer disease management. Medical castration was the first effective systemic targeted therapy for any cancer and, to this day, androgen ablation remains the mainstay of prostate cancer therapy.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Androgen Antagonists , Androgens , Humans , Male , Prostatic Neoplasms/pathology
2.
PLoS One ; 15(1): e0226735, 2020.
Article in English | MEDLINE | ID: mdl-31917811

ABSTRACT

The major milestones in mouse placental development are well described, but our understanding is limited to how the placenta can adapt to damage or changes in the environment. By using stereology and expression of cell cycle markers, we found that the placenta grows under normal conditions not just by hyperplasia of trophoblast cells but also through extensive polyploidy and cell hypertrophy. In response to feeding a low protein diet to mothers prior to and during pregnancy, to mimic chronic malnutrition, we found that this normal program was altered and that it was influenced by the sex of the conceptus. Male fetuses showed intrauterine growth restriction (IUGR) by embryonic day (E) 18.5, just before term, whereas female fetuses showed IUGR as early as E16.5. This difference was correlated with differences in the size of the labyrinth layer of the placenta, the site of nutrient and gas exchange. Functional changes were implied based on up-regulation of nutrient transporter genes. The junctional zone was also affected, with a reduction in both glycogen trophoblast and spongiotrophoblast cells. These changes were associated with increased expression of Phlda2 and reduced expression of Egfr. Polyploidy, which results from endoreduplication, is a normal feature of trophoblast giant cells (TGC) but also spongiotrophoblast cells. Ploidy was increased in sinusoidal-TGCs and spongiotrophoblast cells, but not parietal-TGCs, in low protein placentas. These results indicate that the placenta undergoes a range of changes in development and function in response to poor maternal diet, many of which we interpret are aimed at mitigating the impacts on fetal and maternal health.


Subject(s)
Acclimatization , Diet, Protein-Restricted/adverse effects , Embryo, Mammalian/cytology , Fetal Growth Retardation/etiology , Food Deprivation , Placenta/cytology , Animals , Cell Proliferation , Embryo, Mammalian/physiology , Female , Fetal Development , Fetal Growth Retardation/pathology , Giant Cells , Male , Maternal Nutritional Physiological Phenomena , Mice , Mice, Inbred C57BL , Placenta/physiology , Pregnancy , Trophoblasts/cytology , Trophoblasts/physiology
3.
Nat Rev Urol ; 15(5): 271-286, 2018 05.
Article in English | MEDLINE | ID: mdl-29460922

ABSTRACT

The success of next-generation androgen receptor (AR) pathway inhibitors, such as abiraterone acetate and enzalutamide, in treating prostate cancer has been hampered by the emergence of drug resistance. This acquired drug resistance is driven, in part, by the ability of prostate cancer cells to change their phenotype to adopt AR-independent pathways for growth and survival. Around one-quarter of resistant prostate tumours comprise cells that have undergone cellular reprogramming to become AR-independent and to acquire a continuum of neuroendocrine characteristics. These highly aggressive and lethal tumours, termed neuroendocrine prostate cancer (NEPC), exhibit reactivation of developmental programmes that are associated with epithelial-mesenchymal plasticity and acquisition of stem-like cell properties. In the past few years, our understanding of the link between lineage plasticity and an emergent NEPC phenotype has considerably increased. This new knowledge can contribute to novel therapeutic modalities that are likely to improve the treatment and clinical management of aggressive prostate cancer.


Subject(s)
Androgen Receptor Antagonists/therapeutic use , Cell Plasticity , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/metabolism , Disease Progression , Humans , Male , Phenotype , Prostate/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism
4.
Mol Cell Endocrinol ; 462(Pt A): 17-24, 2018 02 15.
Article in English | MEDLINE | ID: mdl-28315377

ABSTRACT

Recently, there has been renewed interest in the development and characterization of patient-derived tumour xenograft (PDX) models. Numerous PDX models have been established for prostate cancer and, importantly, retain the principal molecular, genetic, and histological characteristics of the donor tumour. As such, these models provide significant improvements over standard cell line xenograft models for biological studies, preclinical drug development, and personalized medicine strategies. This review summarizes the current state of the art in this field, illustrating the opportunities and limitations of PDX models in translational prostate cancer research.


Subject(s)
Prostatic Neoplasms/pathology , Translational Research, Biomedical , Xenograft Model Antitumor Assays , Animals , Biomarkers, Tumor/metabolism , Clinical Trials as Topic , Humans , Male , Precision Medicine
5.
Stem Cells Int ; 2016: 4829602, 2016.
Article in English | MEDLINE | ID: mdl-26880966

ABSTRACT

The therapeutic potential of stem cells relies on dissecting the complex signaling networks that are thought to regulate their pluripotency and self-renewal. Until recently, attention has focused almost exclusively on a small set of "core" transcription factors for maintaining the stem cell state. It is now clear that stem cell regulatory networks are far more complex. In this review, we examine the role of the androgen receptor (AR) in coordinating interactions between signaling nodes that govern the balance of cell fate decisions in prostate stem cells.

6.
Oncotarget ; 6(24): 20570-7, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26011941

ABSTRACT

The triple-negative breast cancer (TNBC) subtype is enriched in cancer stem cells (CSCs) and clinically correlated with the highest rate of recurrence. Several studies implicate the RSK pathway as being pivotal for the growth and proliferation of CSCs, which are postulated to drive tumor relapse. We now address the potential for the newly developed RSK inhibitor LJI308 to target the CSC population and repress TNBC growth and dissemination. Overexpression of the Y-box binding protein-1 (YB-1) oncogene in human mammary epithelial cells (HMECs) drove TNBC tumor formation characterized by a multi-drug resistance phenotype, yet these cells were sensitive to LJI308 in addition to the classic RSK inhibitors BI-D1870 and luteolin. Notably, LJI308 specifically targeted transformed cells as it had little effect on the non-tumorigenic parental HMECs. Loss of cell growth, both in 2D and 3D culture, was attributed to LJI308-induced apoptosis. We discovered CD44+/CD49f+ TNBC cells to be less sensitive to chemotherapy compared to the isogenic CD44-/CD49f- cells. However, inhibition of RSK using LJI308, BI-D1870, or luteolin was sufficient to eradicate the CSC population. We conclude that targeting RSK using specific and potent inhibitors, such as LJI308, delivers the promise of inhibiting the growth of TNBC.


Subject(s)
Drug Resistance, Neoplasm/genetics , Pteridines/metabolism , Triple Negative Breast Neoplasms/genetics , Apoptosis , Cell Line, Tumor , Cell Proliferation , Female , Humans , Neoplastic Stem Cells/pathology , Triple Negative Breast Neoplasms/pathology
7.
Stem Cells ; 32(6): 1437-50, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24648416

ABSTRACT

There is growing evidence that cancer-initiation could result from epigenetic changes. Y-box binding protein-1 (YB-1) is a transcription/translation factor that promotes the formation of tumors in transgenic mice; however, the underlying molecular events are not understood. To explore this in a human model system, YB-1 was expressed in mammary epithelial cells under the control of a tetracycline-inducible promoter. The induction of YB-1 promoted phenotypes associated with malignancy in three-dimensional breast acini cultures. This was attributed to YB-1 enhancing the expression and activity of the histone acetyltransferase p300 leading to chromatin remodeling. Specifically, this relaxation of chromatin allowed YB-1 to bind to the BMI1 promoter. The induction of BMI1 engaged the Polycomb complex resulting in histone H2A ubiquitylation and repression of the CDKN2A locus. These events manifested functionally as enhanced self-renewal capacity that occurred in a BMI1-dependent manner. Conversely, p300 inhibition with anacardic acid prevented YB-1 from binding to the BMI1 promoter and thereby subverted self-renewal. Despite these early changes, full malignant transformation was not achieved until RSK2 became overexpressed concomitant with elevated human telomerase reverse transcriptase (hTERT) activity. The YB-1/RSK2/hTERT expressing cells formed tumors in mice that were molecularly subtyped as basal-like breast cancer. We conclude that YB-1 cooperates with p300 to allow BMI1 to over-ride p16(INK4a) -mediated cell cycle arrest enabling self-renewal and the development of aggressive breast tumors.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast/pathology , Cell Transformation, Neoplastic/metabolism , Chromatin Assembly and Disassembly , Epithelial Cells/metabolism , Y-Box-Binding Protein 1/metabolism , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cellular Reprogramming/genetics , Chromatin Assembly and Disassembly/genetics , E1A-Associated p300 Protein/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Transgenic , Models, Biological , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Transcription, Genetic , Up-Regulation/genetics
8.
Oncotarget ; 2(5): 401-6, 2011 May.
Article in English | MEDLINE | ID: mdl-21576761

ABSTRACT

Surprisingly little is known about the underlying genetic events that trigger the progression of a normal cell into a cancerous cell. We recently developed a YB-1-driven model of pre-malignancy where we uncovered that the oncogene promotes genomic instability through cell cycle checkpoint slippage and centrosome amplification. In this research perspective, we describe a possible mechanism by which YB-1 instigates preneoplastic transformation. Using Kinex antibody microarrays with coverage of 800 proteins, we discovered that pre-malignant cells exhibit deregulated signal transduction along the HER2-MAPK-RSK axis. We will discuss the implications of these finding in regard to early intervention strategies.


Subject(s)
Breast Neoplasms/genetics , Models, Biological , Y-Box-Binding Protein 1/metabolism , Breast Neoplasms/physiopathology , Cell Transformation, Neoplastic/genetics , Disease Progression , Gene Regulatory Networks , Genetic Predisposition to Disease , Humans , MAP Kinase Signaling System/genetics , Molecular Targeted Therapy/trends , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/genetics , Y-Box-Binding Protein 1/genetics
9.
Cancer Res ; 70(7): 2840-51, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20332234

ABSTRACT

Y-box binding protein-1 (YB-1) is an oncogenic transcription/translation factor expressed in >40% of breast cancers, where it is associated with poor prognosis, disease recurrence, and drug resistance. We questioned whether this may be linked to the ability of YB-1 to induce the expression of genes linked to cancer stem cells such as CD44 and CD49f. Herein, we report that YB-1 binds the CD44 and CD49f promoters to transcriptionally upregulate their expressions. The introduction of wild-type (WT) YB-1 or activated P-YB-1(S102) stimulated the production of CD44 and CD49f in MDA-MB-231 and SUM 149 breast cancer cell lines. YB-1-transfected cells also bound to the CD44 ligand hyaluronan more than the control cells. Similarly, YB-1 was induced in immortalized breast epithelial cells and upregulated CD44. Conversely, silencing YB-1 decreased CD44 expression as well as reporter activity in SUM 149 cells. In mice, expression of YB-1 in the mammary gland induces CD44 and CD49f with associated hyperplasia. Further, activated mutant YB-1(S102D) enhances self-renewal, primary and secondary mammosphere growth, and soft-agar colony growth, which were reversible via loss of CD44 or CD49f. We next addressed the consequence of this system on therapeutic responsiveness. Here, we show that paclitaxel induces P-YB-1(S102) expression, nuclear localization of activated YB-1, and CD44 expression. The overexpression of WT YB-1 promotes mammosphere growth in the presence of paclitaxel. Importantly, targeting YB-1 sensitized the CD44(High)/CD24(Low) cells to paclitaxel. In conclusion, YB-1 promotes cancer cell growth and drug resistance through its induction of CD44 and CD49f.


Subject(s)
Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Hyaluronan Receptors/biosynthesis , Integrin alpha6/biosynthesis , Nuclear Proteins/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Growth Processes/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/genetics , Integrin alpha6/genetics , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Paclitaxel/pharmacology , Y-Box-Binding Protein 1
10.
Breast Cancer Res ; 10(6): R99, 2008.
Article in English | MEDLINE | ID: mdl-19036157

ABSTRACT

INTRODUCTION: Basal-like breast cancers (BLBC) frequently overexpress the epidermal growth factor receptor (EGFR) and subsequently have high levels of signaling through the MAP kinase pathway, which is thought to contribute to their aggressive behavior. While we have previously reported the expression of Y-box binding protein-1 (YB-1) in 73% of BLBC, it is unclear whether it can be regulated by a component of the MAP kinase signaling pathway. Phosphorylation of YB-1 at the serine 102 residue is required for transcriptional activation of growth-enhancing genes, such as EGFR. Using Motifscan we identified p90 ribosomal S6 kinase (RSK) as a potential candidate for activating YB-1. METHODS: Inhibition of RSK1 and RSK2 was achieved using siRNA and the small molecule SL0101. RSK1, RSK2, activated RSK and kinase-dead RSK were expressed in HCC1937 cells. Kinase assays were performed to illustrate direct phosphorylation of YB-1 by RSK. The impact of inhibiting RSK on YB-1 function was measured by luciferase assays and chromatin immunoprecipitation. RESULTS: Using an in vitro kinase assay, RSK1 and RSK2 were shown to directly phosphorylate YB-1. Interestingly, they were more effective activators of YB-1 than AKT or another novel YB-1 kinase, PKC alpha. Phosphorylation of YB-1 (serine 102 residue) is blocked by inhibition of the MAP kinase pathway or by perturbing RSK1/RSK2 with siRNA or SL0101. In immortalized breast epithelial cells where RSK is active yet AKT is not, YB-1 is phosphorylated. Supporting this observation, RSK2-/- mouse embryo fibroblasts lose the ability to phosphorylate YB-1 in response to epidermal growth factor. This subsequently interfered with the ability of YB-1 to regulate the expression of EGFR. The RSK inhibitor SL0101 decreased the ability of YB-1 to bind the promoter, transactivate and ultimately reduce EGFR expression. In concordance with these results the expression of constitutively active RSK1 increased YB-1 phosphorylation, yet the kinase-dead RSK did not. CONCLUSIONS: We therefore conclude that RSK1/RSK2 are novel activators of YB-1, able to phosphorylate the serine 102 residue. This provides a newly described mechanism whereby YB-1 is activated in breast cancer. This implicates the EGFR/RSK/YB-1 pathway as an important component of BLBC, providing an important opportunity for therapeutic intervention.


Subject(s)
Breast Neoplasms/metabolism , Neoplasms, Basal Cell/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Y-Box-Binding Protein 1/metabolism , Animals , Benzopyrans/pharmacology , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cells, Cultured , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , ErbB Receptors/metabolism , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , Immunoprecipitation , Luciferases/metabolism , MAP Kinase Signaling System , Mice , Monosaccharides/pharmacology , Neoplasms, Basal Cell/genetics , Neoplasms, Basal Cell/pathology , Phosphorylation/drug effects , Promoter Regions, Genetic , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Serine/chemistry , Serine/metabolism , Y-Box-Binding Protein 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...