42 results on '"Nelson, Colleen C."'
Search Results
2. Disruption of Androgen Regulation in the Prostate by the Environmental Contaminant Hexachlorobenzene
- Author
-
Ralph, Jody L., Orgebin-Crist, Marie-Claire, Lareyre, Jean-Jacques, and Nelson, Colleen C.
- Published
- 2003
3. Therapy-induced lipid uptake and remodeling underpin ferroptosis hypersensitivity in prostate cancer
- Author
-
Tousignant, Kaylyn D., Rockstroh, Anja, Poad, Berwyck L. J., Talebi, Ali, Young, Reuben S. E., Taherian Fard, Atefeh, Gupta, Rajesh, Zang, Tuo, Wang, Chenwei, Lehman, Melanie L., Swinnen, Johan V., Blanksby, Stephen J., Nelson, Colleen C., and Sadowski, Martin C.
- Published
- 2020
- Full Text
- View/download PDF
4. Targeted beta therapy of prostate cancer with 177Lu-labelled Miltuximab® antibody against glypican-1 (GPC-1)
- Author
-
Yeh, Mei-Chun, Tse, Brian W. C., Fletcher, Nicholas L., Houston, Zachary H., Lund, Maria, Volpert, Marianna, Stewart, Chelsea, Sokolowski, Kamil A., Jeet, Varinder, Thurecht, Kristofer J., Campbell, Douglas H., Walsh, Bradley J., Nelson, Colleen C., and Russell, Pamela J.
- Published
- 2020
- Full Text
- View/download PDF
5. Introduction of Androgen Receptor Targeting shRNA Inhibits Tumor Growth in Patient-Derived Prostate Cancer Xenografts.
- Author
-
Thomas, Patrick B., Alinezhad, Saeid, Joshi, Andre, Sweeney, Katrina, Tse, Brian W. C., Tevz, Gregor, McPherson, Stephen, Nelson, Colleen C., Williams, Elizabeth D., and Vela, Ian
- Subjects
ANDROGEN receptors ,GENE targeting ,CANCER cell culture ,TUMOR growth ,PROSTATE cancer ,XENOGRAFTS ,REPORTER genes - Abstract
Patient-derived xenograft (PDX) models have been established as important preclinical cancer models, overcoming some of the limitations associated with the use of cancer cell lines. The utility of prostate cancer PDX models has been limited by an inability to genetically manipulate them in vivo and difficulties sustaining PDX-derived cancer cells in culture. Viable, short-term propagation of PDX models would allow in vitro transfection with traceable reporters or manipulation of gene expression relevant to different studies within the prostate cancer field. Here, we report an organoid culture system that supports the growth of prostate cancer PDX cells in vitro and permits genetic manipulation, substantially increasing the scope to use PDXs to study the pathobiology of prostate cancer and define potential therapeutic targets. We have established a short-term PDX-derived in vitro cell culture system which enables genetic manipulation of prostate cancer PDXs LuCaP35 and BM18. Genetically manipulated cells could be re-established as viable xenografts when re-implanted subcutaneously in immunocompromised mice and were able to be serially passaged. Tumor growth of the androgen-dependent LuCaP35 PDX was significantly inhibited following depletion of the androgen receptor (AR) in vivo. Taken together, this system provides a method to generate novel preclinical models to assess the impact of controlled genetic perturbations and allows for targeting specific genes of interest in the complex biological setting of solid tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Functional localization and competition between the androgen receptor and T-cell factor for nuclear β-catenin: a means for inhibition of the Tcf signaling axis
- Author
-
Mulholland, David J, Read, Jason T, Rennie, Paul S, Cox, Michael E, and Nelson, Colleen C
- Published
- 2003
- Full Text
- View/download PDF
7. Epigenetic Mechanisms for Progression of Prostate Cancer
- Author
-
Rennie, Paul S. and Nelson, Colleen C.
- Published
- 1998
- Full Text
- View/download PDF
8. Leptin antagonism inhibits prostate cancer xenograft growth and progression.
- Author
-
Philp, Lisa K., Rockstroh, Anja, Sadowski, Martin C., Fard, Atefeh Taherian, Lehman, Melanie, Tevz, Gregor, Libério, Michelle S., Bidgood, Charles L., Gunter, Jennifer H., McPherson, Stephen, Bartonicek, Nenad, Wade, John D., Otvos Jr., Laszlo, and Nelson, Colleen C.
- Subjects
LEPTIN ,ANDROGEN receptors ,TUMOR growth ,PROSTATE cancer ,DRUG side effects ,ANDROGEN deprivation therapy ,LEPTIN receptors - Abstract
Hyperleptinaemia is a well-established therapeutic side effect of drugs inhibiting the androgen axis in prostate cancer (PCa), including main stay androgen deprivation therapy (ADT) and androgen targeted therapies (ATT). Given significant crossover between the adipokine hormone signalling of leptin and multiple cancer-promoting hallmark pathways, including growth, proliferation, migration, angiogenesis, metabolism and inflammation, targeting the leptin axis is therapeutically appealing, especially in advanced PCa where current therapies fail to be curative. In this study, we uncover leptin as a novel universal target in PCa and are the first to highlight increased intratumoural leptin and leptin receptor (LEPR) expression in PCa cells and patients' tumours exposed to androgen deprivation, as is observed in patients' tumours of metastatic and castrate resistant (CRPC) PCa. We also reveal the world-first preclinical evidence that demonstrates marked efficacy of targeted leptin-signalling blockade, using Allo-aca, a potent, specific, and safe LEPR peptide antagonist. Allo-aca-suppressed tumour growth and delayed progression to CRPC in mice bearing LNCaP xenografts, with reduced tumour v ascularity and altered pathways of apoptosis, transcription/translation, and energetics in tumours determined as potential mechanisms underpinning anti-tumour efficacy. We highlight LEPR blockade in combination with androgen axis inhibition represents a promising new therapeutic strategy vital in advanced PCa treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
9. Insulin Enhances Migration and Invasion in Prostate Cancer Cells by Up-Regulation of FOXC2.
- Author
-
Sarkar, Phoebe L., Lee, Wendy, Williams, Elizabeth D., Lubik, Amy A., Stylianou, Nataly, Shokoohmand, Ali, Lehman, Melanie L., Hollier, Brett G., Gunter, Jennifer H., and Nelson, Colleen C.
- Subjects
CANCER cell motility ,CANCER cells ,INSULIN ,PROSTATE cancer ,INSULIN receptors ,TRANSCRIPTION factors ,FORKHEAD transcription factors ,ABIRATERONE acetate - Abstract
Androgen deprivation therapy (ADT) is the standard treatment for advanced prostate cancer (PCa), yet many patients relapse with lethal metastatic disease. With this loss of androgens, increased cell plasticity has been observed as an adaptive response to ADT. This includes gain of invasive and migratory capabilities, which may contribute to PCa metastasis. Hyperinsulinemia, which develops as a side-effect of ADT, has been associated with increased tumor aggressiveness and faster treatment failure. We investigated the direct effects of insulin in PCa cells that may contribute to this progression. We measured cell migration and invasion induced by insulin using wound healing and transwell assays in a range of PCa cell lines of variable androgen dependency (LNCaP, 22RV1, DuCaP, and DU145 cell lines). To determine the molecular events driving insulin-induced invasion we used transcriptomics, quantitative real time-PCR, and immunoblotting in three PCa cell lines. Insulin increased invasiveness of PCa cells, upregulating Forkhead Box Protein C2 (FOXC2), and activating key PCa cell plasticity mechanisms including gene changes consistent with epithelial-to-mesenchymal transition (EMT) and a neuroendocrine phenotype. Additionally, analysis of publicly available clinical PCa tumor data showed metastatic prostate tumors demonstrate a positive correlation between insulin receptor expression and the EMT transcription factor FOXC2. The insulin receptor is not suitable to target clinically however, our data shows that actions of insulin in PCa cells may be suppressed by inhibiting downstream signaling molecules, PI3K and ERK1/2. This study identifies for the first time, a mechanism for insulin-driven cancer cell motility and supports the concept that targeting insulin signaling at the level of the PCa tumor may extend the therapeutic efficacy of ADT. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
10. Limited short-term effects on human prostate cancer xenograft growth and epidermal growth factor receptor gene expression by the ghrelin receptor antagonist [D-Lys3]-GHRP-6.
- Author
-
Maugham, Michelle L., Seim, Inge, Thomas, Patrick B., Crisp, Gabrielle J., Shah, Esha T., Herington, Adrian C., Gregory, Laura S., Nelson, Colleen C., Jeffery, Penny L., and Chopin, Lisa K.
- Abstract
Purpose: The ghrelin axis regulates many physiological functions (including appetite, metabolism, and energy balance) and plays a role in disease processes. As ghrelin stimulates prostate cancer proliferation, the ghrelin receptor antagonist [D-Lys
3 ]-GHRP-6 is a potential treatment for castrate-resistant prostate cancer and for preventing the metabolic consequences of androgen-targeted therapies. We therefore explored the effect of [D-Lys3 ]-GHRP-6 on PC3 prostate cancer xenograft growth. Methods: NOD/SCID mice with PC3 prostate cancer xenografts were administered 20 nmoles/mouse [D-Lys3 ]-GHRP-6 daily by intraperitoneal injection for 14 days and tumour volume and weight were measured. RNA sequencing of tumours was conducted to investigate expression changes following [D-Lys3 ]-GHRP-6 treatment. A second experiment, extending treatment time to 18 days and including a higher dose of [D-Lys3 ]-GHRP-6 (200 nmoles/mouse/day), was undertaken to ensure repeatability. Results: We demonstrate here that daily intraperitoneal injection of 20 nmoles/mouse [D-Lys3 ]-GHRP-6 reduces PC3 prostate cancer xenograft tumour volume and weight in NOD/SCID mice at two weeks post treatment initiation. RNA-sequencing revealed reduced expression of epidermal growth factor receptor (EGFR) in these tumours. Further experiments demonstrated that the effects of [D-Lys3 ]-GHRP-6 are transitory and lost after 18 days of treatment. Conclusions: We show that [D-Lys3 ]-GHRP-6 has transitory effects on prostate xenograft tumours in mice, which rapidly develop an apparent resistance to the antagonist. Although further studies on [D-Lys3 ]-GHRP-6 are warranted, we suggest that daily treatment with the antagonist is not a suitable treatment for advanced prostate cancer. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
11. No effect of unacylated ghrelin administration on subcutaneous PC3 xenograft growth or metabolic parameters in a Rag1-/- mouse model of metabolic dysfunction.
- Author
-
Maugham, Michelle L., Seim, Inge, Thomas, Patrick B., Crisp, Gabrielle J., Shah, Esha T., Herington, Adrian C., Brown, Kristy A., Gregory, Laura S., Nelson, Colleen C., Jeffery, Penny L., and Chopin, Lisa K.
- Subjects
GHRELIN ,XENOGRAFTS ,HIGH-fat diet ,METABOLIC syndrome ,PROSTATE cancer - Abstract
Ghrelin is a peptide hormone which, when acylated, regulates appetite, energy balance and a range of other biological processes. Ghrelin predominately circulates in its unacylated form (unacylated ghrelin; UAG). UAG has a number of functions independent of acylated ghrelin, including modulation of metabolic parameters and cancer progression. UAG has also been postulated to antagonise some of the metabolic effects of acyl-ghrelin, including its effects on glucose and insulin regulation. In this study, Rag1
-/- mice with high-fat diet-induced obesity and hyperinsulinaemia were subcutaneously implanted with PC3 prostate cancer xenografts to investigate the effect of UAG treatment on metabolic parameters and xenograft growth. Daily intraperitoneal injection of 100 μg/kg UAG had no effect on xenograft tumour growth in mice fed normal rodent chow or 23% high-fat diet. UAG significantly improved glucose tolerance in host Rag1-/- mice on a high-fat diet, but did not significantly improve other metabolic parameters. We propose that UAG is not likely to be an effective treatment for prostate cancer, with or without associated metabolic syndrome. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
12. Mining human cancer datasets for kallikrein expression in cancer: the 'KLK-CANMAP' Shiny web tool.
- Author
-
Chenwei Wang, Moya, Leire, Clements, Judith A., Nelson, Colleen C., and Batra, Jyotsna
- Subjects
KALLIKREIN ,CANCER cells ,CANCER treatment ,GENE expression ,PROSTATE cancer ,BIOLOGICAL tags - Abstract
The dysregulation of the serine-protease family kallikreins (KLKs), comprising 15 genes, has been reportedly associated with cancer. Their expression in several tissues and physiological fluids makes them potential candidates as biomarkers and therapeutic targets. There are several databases available to mine gene expression in cancer, which often include clinical and pathological data. However, these platforms present some limitations when comparing a specific set of genes and can generate considerable unwanted data. Here, several datasets that showed significant differential expression (p < 0.01) in cancer vs. normal (n = 118), metastasis vs. primary (n = 15) and association with cancer survival (n = 21) have been compiled in a user-friendly format from two open and/or publicly available databases Oncomine and OncoLnc for the 15 KLKs. The data have been included in a free web application tool: the KLK-CANMAP https://cancerbioinformatics. shinyapps.io/klk-canmap/. This tool integrates, analyses and visualises data and it was developed with the R Shiny framework. Using KLK-CANMAP box-plots, heatmaps and Kaplan-Meier graphs can be generated for the KLKs of interest. We believe this new cancer KLK focused web tool will benefit the KLK community by narrowing the data visualisation to only the genes of interest. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
13. Discovery of thalicthuberine as a novel antimitotic agent from nature that disrupts microtubule dynamics and induces apoptosis in prostate cancer cells.
- Author
-
Levrier, Claire, Rockstroh, Anja, Gabrielli, Brian, Kavallaris, Maria, Lehman, Melanie, Davis, Rohan A., Sadowski, Martin C., and Nelson, Colleen C.
- Abstract
We report for the first time the mechanism of action of the natural product thalicthuberine (TH) in prostate and cervical cancer cells. TH induced a strong accumulation of LNCaP cells in mitosis, severe mitotic spindle defects, and asymmetric cell divisions, ultimately leading to mitotic catastrophe accompanied by cell death through apoptosis. However, unlike microtubule-binding drugs (vinblastine and paclitaxel), TH did not directly inhibit tubulin polymerization when tested in a cell-free system, whereas it reduced cellular microtubule polymer mass in LNCaP cells. This suggests that TH indirectly targets microtubule dynamics through inhibition of a critical regulator or tubulin-associated protein. Furthermore, TH is not a major substrate for P-glycoprotein (Pgp), which is responsible for multidrug resistance in numerous cancers, providing a rationale to further study TH in cancers with Pgp-mediated treatment resistance. The identification of TH's molecular target in future studies will be of great value to the development of TH as potential treatment of multidrug-resistant tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
14. Fusion transcript loci share many genomic features with non-fusion loci.
- Author
-
Lai, John, Jiyuan An, Seim, Inge, Walpole, Carina, Hoffman, Andrea, Moya, Leire, Srinivasan, Srilakshmi, Perry-Keene, Joanna L., Chenwei Wang, Lehman, Melanie L., Nelson, Colleen C., Clements, Judith A., and Batra, Jyotsna
- Subjects
NUCLEOTIDE sequencing ,PROSTATE cancer ,NUCLEOTIDE sequence ,ANDROGEN receptors ,REGENERATION (Biology) - Abstract
Background: Fusion transcripts are found in many tissues and have the potential to create novel functional products. Here, we investigate the genomic sequences around fusion junctions to better understand the transcriptional mechanisms mediating fusion transcription/splicing. We analyzed data from prostate (cancer) cells as previous studies have shown extensively that these cells readily undergo fusion transcription. Results: We used the FusionMap program to identify high-confidence fusion transcripts from RNAseq data. The RNAseq datasets were from our (N = 8) and other (N = 14) clinical prostate tumors with adjacent non-cancer cells, and from the LNCaP prostate cancer cell line that were mock-, androgen- (DHT), and anti-androgen- (bicalutamide, enzalutamide) treated. In total, 185 fusion transcripts were identified from all RNAseq datasets. The majority (76 %) of these fusion transcripts were 'read-through chimeras' derived from adjacent genes in the genome. Characterization of sequences at fusion loci were carried out using a combination of the FusionMap program, custom Perl scripts, and the RNAfold program. Our computational analysis indicated that most fusion junctions (76 %) use the consensus GT-AG intron donor-acceptor splice site, and most fusion transcripts (85 %) maintained the open reading frame. We assessed whether parental genes of fusion transcripts have the potential to form complementary base pairing between parental genes which might bring them into physical proximity. Our computational analysis of sequences flanking fusion junctions at parental loci indicate that these loci have a similar propensity as non-fusion loci to hybridize. The abundance of repetitive sequences at fusion and non-fusion loci was also investigated given that SINE repeats are involved in aberrant gene transcription. We found few instances of repetitive sequences at both fusion and non-fusion junctions. Finally, RT-qPCR was performed on RNA from both clinical prostate tumors and adjacent non-cancer cells (N = 7), and LNCaP cells treated as above to validate the expression of seven fusion transcripts and their respective parental genes. We reveal that fusion transcript expression is similar to the expression of parental genes. Conclusions: Fusion transcripts maintain the open reading frame, and likely use the same transcriptional machinery as non-fusion transcripts as they share many genomic features at splice/fusion junctions. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
15. Targeting ASCT2-mediated glutamine uptake blocks prostate cancer growth and tumour development.
- Author
-
Wang, Qian, Hardie, Rae‐Anne, Hoy, Andrew J, van Geldermalsen, Michelle, Gao, Dadi, Fazli, Ladan, Sadowski, Martin C, Balaban, Seher, Schreuder, Mark, Nagarajah, Rajini, Wong, Justin J‐L, Metierre, Cynthia, Pinello, Natalia, Otte, Nicholas J, Lehman, Melanie L, Gleave, Martin, Nelson, Colleen C, Bailey, Charles G, Ritchie, William, and Rasko, John EJ
- Abstract
Glutamine is conditionally essential in cancer cells, being utilized as a carbon and nitrogen source for macromolecule production, as well as for anaplerotic reactions fuelling the tricarboxylic acid ( TCA) cycle. In this study, we demonstrated that the glutamine transporter ASCT2 ( SLC1A5) is highly expressed in prostate cancer patient samples. Using LNCaP and PC-3 prostate cancer cell lines, we showed that chemical or shRNA-mediated inhibition of ASCT2 function in vitro decreases glutamine uptake, cell cycle progression through E2F transcription factors, mTORC1 pathway activation and cell growth. Chemical inhibition also reduces basal oxygen consumption and fatty acid synthesis, showing that downstream metabolic function is reliant on ASCT2-mediated glutamine uptake. Furthermore, shRNA knockdown of ASCT2 in PC-3 cell xenografts significantly inhibits tumour growth and metastasis in vivo, associated with the down-regulation of E2F cell cycle pathway proteins. In conclusion, ASCT2-mediated glutamine uptake is essential for multiple pathways regulating the cell cycle and cell growth, and is therefore a putative therapeutic target in prostate cancer. © 2015 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
16. Adverse effects of androgen-deprivation therapy in prostate cancer and their management.
- Author
-
Rhee, Handoo, Gunter, Jennifer H., Heathcote, Peter, Ho, Ken, Stricker, Phillip, Corcoran, Niall M., and Nelson, Colleen C.
- Subjects
PROSTATE cancer ,DRUG side effects ,CARDIOVASCULAR diseases ,ANDROGEN drugs ,CASTRATION - Abstract
Objective To provide an up-to-date summary of current literature on the management of adverse effects of androgen-deprivation therapy (ADT). Patients and Methods All relevant medical literature on men with prostate cancer treated with ADT from 2005 to 2014, and older relevant papers, were reviewed. Recent health advisory statements from the Australian government, societies and advocacy groups have been incorporated to the document. Results There are numerous adverse effects of ADT that require pro-active prevention and treatment. Ranging from cardiovascular disease, diabetes and osteoporosis, to depression, cognitive decline and sexual dysfunction, the range of adverse effects is wide. Baseline assessment, monitoring, prevention and consultation from a multidisciplinary team are important in minimising the harm from ADT. Conclusions This review provides a series of practical recommendations to assist with managing the adverse effects of ADT. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
17. The tumour-promoting receptor tyrosine kinase, EphB4, regulates expression of Integrin-β8 in prostate cancer cells.
- Author
-
Mertens-Walker, Inga, Fernandini, Bruno C., Maharaj, Mohanan S. N., Rockstroh, Anja, Nelson, Colleen C., Herington, Adrian C., and Stephenson, Sally-Anne
- Subjects
PROSTATE cancer ,CANCER invasiveness ,PROTEIN-tyrosine kinases ,COCARCINOGENS ,CANCER cells ,INTEGRINS ,MICROARRAY technology - Abstract
Background: The EphB4 receptor tyrosine kinase is overexpressed in many cancers including prostate cancer. The molecular mechanisms by which this ephrin receptor influences cancer progression are complex as there are tumor-promoting ligand-independent mechanisms in place as well as ligand-dependent tumor suppressive pathways. Methods: We employed transient knockdown of EPHB4 in prostate cancer cells, coupled with gene microarray analysis, to identify genes that were regulated by EPHB4 and may represent linked tumor-promoting factors. We validated target genes using qRT-PCR and employed functional assays to determine their role in prostate cancer migration and invasion. Results: We discovered that over 500 genes were deregulated upon EPHB4 siRNA knockdown, with integrin β8 (ITGB8) being the top hit (29-fold down-regulated compared to negative non-silencing siRNA). Gene ontology analysis found that the process of cell adhesion was highly deregulated and two other integrin genes, ITGA3 and ITGA10, were also differentially expressed. In parallel, we also discovered that over-expression of EPHB4 led to a concomitant increase in ITGB8 expression. In silico analysis of a prostate cancer progression microarray publically available in the Oncomine database showed that both EPHB4 and ITGB8 are highly expressed in prostatic intraepithelial neoplasia, the precursor to prostate cancer. Knockdown of ITGB8 in PC-3 and 22Rv1 prostate cancer cells in vitro resulted in significant reduction of cell migration and invasion. Conclusions: These results reveal that EphB4 regulates integrin β8 expression and that integrin β8 plays a hitherto unrecognized role in the motility of prostate cancer cells and thus targeting integrin β8 may be a new treatment strategy for prostate cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
18. Androgen-targeted therapy-induced epithelial mesenchymal plasticity and neuroendocrine transdifferentiation in prostate cancer: an opportunity for intervention.
- Author
-
Nouri, Mannan, Ratther, Ellca, Stylianou, Nataly, Nelson, Colleen C., Hollier, Brett G., and Williams, Elizabeth D.
- Subjects
PROSTATE cancer treatment ,ANDROGEN drugs ,CELL differentiation ,PHENOTYPIC plasticity ,NEUROENDOCRINOLOGY ,MESENCHYMAL stem cells - Abstract
Androgens regulate biological pathways to promote proliferation, differentiation, and survival of benign and malignant prostate tissue. Androgen receptor (AR) targeted therapies exploit this dependence and are used in advanced prostate cancer to control disease progression. Contemporary treatment regimens involve sequential use of inhibitors of androgen synthesis or AR function. Although targeting the androgen axis has clear therapeutic benefit, its effectiveness is temporary, as prostate tumor cells adapt to survive and grow. The removal of androgens (androgen deprivation) has been shown to activate both epithelial-to-mesenchymal transition (EMT) and neuroendocrine transdifferentiation (NEtD) programs. EMT has established roles in promoting biological phenotypes associated with tumor progression (migration/invasion, tumor cell survival, cancer stem cell-like properties, resistance to radiation and chemotherapy) in multiple human cancer types. NEtD in prostate cancer is associated with resistance to therapy, visceral metastasis, and aggressive disease. Thus, activation of these programs via inhibition of the androgen axis provides a mechanism by which tumor cells can adapt to promote disease recurrence and progression. Brachyury, Axl, MEK, and Aurora kinase A are molecular drivers of these programs, and inhibitors are currently in clinical trials to determine therapeutic applications. Understanding tumor cell plasticity will be important in further defining the rational use of androgen-targeted therapies clinically and provides an opportunity for intervention to prolong survival of men with metastatic prostate cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
19. Differential Effects of Tissue Culture Coating Substrates on Prostate Cancer Cell Adherence, Morphology and Behavior.
- Author
-
Liberio, Michelle S., Sadowski, Martin C., Soekmadji, Carolina, Davis, Rohan A., and Nelson, Colleen C.
- Subjects
PROSTATE cancer ,TISSUE culture ,CELL lines ,CELL membranes ,CELL-matrix adhesions ,SURFACE coatings - Abstract
Weak cell-surface adhesion of cell lines to tissue culture surfaces is a common problem and presents technical limitations to the design of experiments. To overcome this problem, various surface coating protocols have been developed. However, a comparative and precise real-time measurement of their impact on cell behavior has not been conducted. The prostate cancer cell line LNCaP, derived from a patient lymph node metastasis, is a commonly used model system in prostate cancer research. However, the cells’ characteristically weak attachment to the surface of tissue culture vessels and cover slips has impeded their manipulation and analysis and use in high throughput screening. To improve the adherence of LNCaP cells to the culture surface, we compared different coating reagents (poly--lysine, poly--ornithine, collagen type IV, fibronectin, and laminin) and culturing conditions and analyzed their impact on cell proliferation, adhesion, morphology, mobility and gene expression using real-time technologies. The results showed that fibronectin, poly--lysine and poly--ornithine improved LNCaP cells adherence and provoked cell morphology alterations, such as increase of nuclear and cellular area. These coating reagents also induced a higher expression of F-actin and reduced cell mobility. In contrast, laminin and collagen type IV did not improve adherence but promoted cell aggregation and affected cell morphology. Cells cultured in the presence of laminin displayed higher mobility than control cells. All the coating conditions significantly affected cell viability; however, they did not affect the expression of androgen receptor-regulated genes. Our comparative findings provide important insight for the selection of the ideal coating reagent and culture conditions for the cancer cell lines with respect to their effect on proliferation rate, attachment, morphology, migration, transcriptional response and cellular cytoskeleton arrangement. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
20. Analysis of androgen and anti-androgen regulation of KLK-related peptidase 2, 3, and 4 alternative transcripts in prostate cancer.
- Author
-
Lai, John, Jiyuan An, Nelson, Colleen C., Lehman, Melanie L., Batra, Jyotsna, and Clements, Judith A.
- Subjects
ANDROGEN receptors ,ANTIANDROGENS ,KALLIKREIN ,PEPTIDASE ,PROSTATE cancer ,STANOLONE - Abstract
We assessed whether alternative transcripts (using KLK2, KLK3 and KLK4 as models) are differentially regulated by androgens and anti-androgens as an indicator of prostate cancers as they acquire treatment resistance. Using RNAseq of LNCaP cells treated with dihydrotestosterone, bicalutamide and enzalutamide, we show that the expression of variant KLK transcripts is markedly different to other variant transcripts at those loci. We also reveal that KLK variants are also over 2-fold more highly expressed in prostate cancers compared to their corresponding normal prostate. We propose that androgens and anti-androgens can activate specific variant transcripts of critical prostate cancer genes during treatment resistance. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
21. Exosomes in Prostate Cancer: Putting Together the Pieces of a Puzzle.
- Author
-
Soekmadji, Carolina, Russell, Pamela J., and Nelson, Colleen C.
- Subjects
BIOMARKERS ,CYTOPLASM ,PROSTATE tumors ,PHYSIOLOGY - Abstract
Exosomes have been shown to act as mediators for cell to cell communication and as a potential source of biomarkers for many diseases, including prostate cancer. Exosomes are nanosized vesicles secreted by cells and consist of proteins normally found in multivesicular bodies, RNA, DNA and lipids. As a potential source of biomarkers, exosomes have attracted considerable attention, as their protein content resembles that of their cells of origin, even though it is noted that the proteins, miRNAs and lipids found in the exosomes are not a reflective stoichiometric sampling of the contents from the parent cells. While the biogenesis of exosomes in dendritic cells and platelets has been extensively characterized, much less is known about the biogenesis of exosomes in cancer cells. An understanding of the processes involved in prostate cancer will help to further elucidate the role of exosomes and other extracellular vesicles in prostate cancer progression and metastasis. There are few methodologies available for general isolation of exosomes, however validation of those methodologies is necessary to study the role of exosomal-derived biomarkers in various diseases. In this review, we discuss "exosomes" as a member of the family of extracellular vesicles and their potential to provide candidate biomarkers for prostate cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
22. Targeting Amino Acid Transport in Metastatic Castration-Resistant Prostate Cancer: Effects on Cell Cycle, Cell Growth, and Tumor Development.
- Author
-
Wang, Qian, Tiffen, Jessamy, Bailey, Charles G., Lehman, Melanie L., Ritchie, William, Fazli, Ladan, Metierre, Cynthia, Feng, Yue (Julie), Li, Estelle, Gleave, Martin, Buchanan, Grant, Nelson, Colleen C., Rasko, John E. J., and Holst, Jeff
- Subjects
AMINO acids ,PHYSIOLOGICAL effects of leucine ,CANCER cells ,PROSTATE cancer ,TRANSCRIPTION factors - Abstract
Background L-type amino acid transporters (LATs) uptake neutral amino acids including L-leucine into cells, stimulating mammalian target of rapamycin complex 1 signaling and protein synthesis. LAT1 and LAT3 are overexpressed at different stages of prostate cancer, and they are responsible for increasing nutrients and stimulating cell growth. Methods We examined LAT3 protein expression in human prostate cancer tissue microarrays. LAT function was inhibited using a leucine analog (BCH) in androgen-dependent and -independent environments, with gene expression analyzed by microarray. A PC-3 xenograft mouse model was used to study the effects of inhibiting LAT1 and LAT3 expression. Results were analyzed with the Mann-Whitney U or Fisher exact tests. All statistical tests were two-sided. Results LAT3 protein was expressed at all stages of prostate cancer, with a statistically significant decrease in expression after 4–7 months of neoadjuvant hormone therapy (4–7 month mean = 1.571; 95% confidence interval = 1.155 to 1.987 vs 0 month = 2.098; 95% confidence interval = 1.962 to 2.235; P = .0187). Inhibition of LAT function led to activating transcription factor 4–mediated upregulation of amino acid transporters including ASCT1, ASCT2, and 4F2hc, all of which were also regulated via the androgen receptor. LAT inhibition suppressed M-phase cell cycle genes regulated by E2F family transcription factors including critical castration-resistant prostate cancer regulatory genes UBE2C, CDC20, and CDK1. In silico analysis of BCH-downregulated genes showed that 90.9% are statistically significantly upregulated in metastatic castration-resistant prostate cancer. Finally, LAT1 or LAT3 knockdown in xenografts inhibited tumor growth, cell cycle progression, and spontaneous metastasis in vivo. Conclusion Inhibition of LAT transporters may provide a novel therapeutic target in metastatic castration-resistant prostate cancer, via suppression of mammalian target of rapamycin complex 1 activity and M-phase cell cycle genes. [ABSTRACT FROM PUBLISHER]
- Published
- 2013
- Full Text
- View/download PDF
23. IGF2 increases de novo steroidogenesis in prostate cancer cells.
- Author
-
Lubik, Amy A., Gunter, Jennifer H., Hollier, Brett G., Ettinger, Susan, Fazli, Ladan, Stylianou, Nataly, Hendy, Stephen C., Adomat, Hans H., Gleave, Martin E., Pollak, Michael, Herington, Adrian, and Nelson, Colleen C.
- Subjects
CANCER patients ,PROSTATE cancer ,PEPTIDES ,IMMUNOGLOBULINS ,THERAPEUTICS - Abstract
IGF2 is a mitogenic foetal growth factor commonly over-expressed in cancers, including prostate cancer (PC). We recently demonstrated that insulin can activate de novo steroidogenesis in PC cells, a major pathway for reactivation of androgen pathways and PC progression. IGF2 can activate the IGF1 receptor (IGF1R) or insulin receptor (INSR) or hybrids of these two receptors. We therefore hypothesized that IGF2 may contribute to PC progression via de novo steroidogenesis. IGF2 mRNA but not IGF2 receptor mRNA expression was increased in patient samples during progression to castrate-resistant PC as was immunoreactivity to INSR and IGF1R antibodies. Treatment of androgen receptor (AR)-positive PC cell lines LNCaP and 22RV1 with IGF2 for 48 h resulted in increased expression of steroidogenic enzyme mRNA and protein, including steroid acute regulatory protein (StAR), cytochrome p450 family member (CYP)17A1, aldo-keto reductase family member (AKR)1C3 and hydroxysteroid dehydrogenase (HSD)17B3. IGF2 treatment resulted in increased steady state steroid levels and increased de novo steroidogenesis resulting in AR activation as demonstrated by PSA mRNA induction. Inhibition of the IGF1R/INSR signalling axis attenuated the effects of IGF2 on steroid hormone synthesis. We present a potential mechanism for prostatic IGF2 contributing to PC progression by inducing steroidogenesis and that IGF2 signalling and related pathways present attractive targets for PC therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
24. A gene signature identified using a mouse model of androgen receptor-dependent prostate cancer predicts biochemical relapse in human disease.
- Author
-
Thompson, Vanessa C., Day, Tanya K., Bianco-Miotto, Tina, Selth, Luke A., Han, Guangzhou, Thomas, Mervyn, Buchanan, Grant, Scher, Howard I., Nelson, Colleen C., Greenberg, Norman M., Butler, Lisa M., and Tilley, Wayne D.
- Abstract
Mutations in the androgen receptor (AR) have been detected in experimental and clinical prostate tumors. Mice with enforced prostate-specific expression of one such receptor variant, AR-E231G, invariably develop prostatic intraepithelial neoplasia by 12 weeks and metastatic prostate cancer by 52 weeks. The aim of this study was to identify genes with altered expression in the prostates of AR-E231G mice at an early stage of disease that may act as drivers of AR-mediated tumorigenesis. The gene expression profile of AR-E231G prostate tissue from 12-week-old mice was compared to an equivalent profile from mice expressing the AR-T857A receptor variant (analogous to the AR-T877A variant in LNCaP cells), which do not develop prostate tumors. One hundred and thirty-two genes were differentially expressed in AR-E231G prostates. Classification of these genes revealed enrichment for cellular pathways known to be involved in prostate cancer, including cell cycle and lipid metabolism. Suppression of two genes upregulated in the AR-E231G model, ADM and CITED1, increased cell death and reduced proliferation of human prostate cancer cells. Many genes differentially expressed in AR-E231G prostates are also deregulated in human tumors. Three of these genes, ID4, NR2F1 and PTGDS, which were expressed at consistently lower levels in clinical prostate cancer compared to nonmalignant tissues, formed a signature that predicted biochemical relapse (hazard ratio 2.2, p = 0.038). We believe that our findings support the value of this novel mouse model of prostate cancer to identify candidate therapeutic targets and/or biomarkers of human disease. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
25. Steroidogenesis inhibitors alter but do not eliminate androgen synthesis mechanisms during progression to castration-resistance in LNCaP prostate xenografts
- Author
-
Locke, Jennifer A., Nelson, Colleen C., Adomat, Hans H., Hendy, Stephen C., Gleave, Martin E., and Guns, Emma S. Tomlinson
- Subjects
- *
HORMONE synthesis , *LIPID metabolism , *ANDROGENS , *REACTION mechanisms (Chemistry) , *CASTRATION , *PROSTATE cancer , *XENOGRAFTS - Abstract
Abstract: In castration-resistant prostate cancer (CRPC) many androgen-regulated genes become re-expressed and tissue androgen levels increase despite low serum levels. We and others have recently reported that CRPC tumor cells can de novo synthesize androgens from adrenal steroid precursors or cholesterol and that high levels of progesterone exist in LNCaP tumors after castration serving perhaps as an intermediate in androgen synthesis. Herein, we compare androgen synthesis from [3H-progesterone] in the presence of specific steroidogenesis inhibitors and anti-androgens in steroid starved LNCaP cells and CRPC tumors. Similarly, we compare steroid profiles in LNCaP tumors at different stages of CRPC progression. Steroidogenesis inhibitors targeting CYP17A1 and SRD5A2 significantly altered but did not eliminate androgen synthesis from progesterone in steroid starved LNCaP cells and CRPC tumors. Upon exposure to inhibitors of steroidogenesis prostate cancer cells adapt gradually during CRPC progression to synthesize DHT in a compensatory manner through alternative feed-forward mechanisms. Furthermore, tumors obtained immediately after castration are significantly less efficient at metabolizing progesterone (∼36%) and produce a different steroid profile to CRPC tumors. Optimal targeting of the androgen axis may be most effective when tumors are least efficient at synthesizing androgens. Confirmatory studies in humans are required to validate these findings. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
26. Comprehensive expression analysis of l-dopa decarboxylase and established neuroendocrine markers in neoadjuvant hormone-treated versus varying Gleason grade prostate tumors.
- Author
-
Wafa, Latif A., Palmer, Jodie, Fazli, Ladan, Hurtado-Coll, Antonio, Bell, Robert H., Nelson, Colleen C., Gleave, Martin E., Cox, Michael E., and Rennie, Paul S.
- Subjects
PROSTATE cancer ,CANCER hormone therapy ,IMMUNOCYTOCHEMISTRY ,TUMOR growth - Abstract
Summary: Current hormone withdrawal therapies used for treatment of advanced prostate cancer lead to androgen-independent tumor growth. Increased prostatic neuroendocrine (NE) cell density has been implicated in promoting progression of prostate cancer, but the process by which this occurs remains unclear. The aim of this study was to determine whether there is an association of increased NE differentiation with neoadjuvant hormone therapy and Gleason grade. Using adjacently sectioned tissue microarrays, the expression profile of novel and known NE markers were monitored. l-Dopa decarboxylase (DDC), a catecholamine synthesis enzyme and androgen receptor (AR) coregulator protein, was identified as an additional NE marker of prostate cancer. Immunohistochemical analysis of DDC with the established NE markers, chromogranin A and bombesin, revealed a significant increase in NE differentiation after 6 months of hormone therapy and after progression to androgen independence but no apparent correlation with Gleason grade. In addition, dual immunofluorescence analysis revealed that approximately 55% of the mixed population of DDC- and chromogranin A-expressing NE cells continue to express AR. Taken together, these results suggest that the increase of NE differentiation in prostate cancers depends specifically on duration of hormone therapy. This increase may be due to the transdifferentiation of AR-expressing epithelial-derived adenocarcinoma cells into an NE cell phenotype. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
27. Demonstration of Upregulated H2 Relaxin mRNA Expression during Neuroendocrine Differentiation of LNCaP Prostate Cancer Cells and Production of Biologically Active Mammalian Recombinant 6 Histidine-Tagged H2 Relaxin.
- Author
-
FIGUEIREDO, KEVIN A., PALMER, JODIE B., MUI, ALICE L., NELSON, COLLEEN C., and COX, MICHAEL E.
- Subjects
RELAXIN ,PROSTATE cancer ,BREAST cancer ,NEUROENDOCRINE tumors ,MACROPHAGES - Abstract
Relaxin was recently implicated as a regulator of breast and prostate cancer progression. We characterized upregulated H2 relaxin gene expression during neuroendocrine differentiation of the human prostate cancer model, LNCaP. To examine the impact of relaxin on host cells associated with prostatic adenocarcinomas, we generated recombinant 6 His-tagged relaxin (RLXH) in a mammalian expression system. This immunoreactive and biologically active relaxin preparation was used to screen a variety of cell types for cAMP responsiveness. Of the cell types screened, none was more responsive to RLXH than the well-characterized monocyte/macrophage cell line THP-1. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
28. Functional localization and competition between the androgen receptor and T-cell factor for nuclear ß-catenin: a means for inhibition of the Tcf signaling axis.
- Author
-
Mulholland, David J, Read, Jason T, Rennie, Paul S, Cox, Michael E, and Nelson, Colleen C
- Subjects
T cells ,PROSTATE cancer ,ANDROGENS ,COLON cancer ,TUMORS - Abstract
Recent reports suggest that the ß-catenin-T-cell factor (Tcf) (BCT) signaling pathway is important in the progression of prostate cancer. Evidence suggests that the androgen receptor (AR) can repress BCT-mediated transcription both in prostate cancer and colon cancer cells (Chesire and Isaacs, 2002). In this study, we validate such findings and show that repression of BCT signaling is facilitated by competition between the AR and Tcf. Measurements of the Tcf transcriptional reporter (TOPFLASH) indicated that AR+DHT-mediated repression can inhibit BCT transcription in the presence of WT and exogenous activating ß-catenin (?1-130?bp). Transient transfections in SW480 cells (APC
mut/mut ) showed that this mode of repression is functionally independent of APC-mediated ß-catenin ubiquitination. Using a recently developed red flourescent protein (HcRed), we demonstrate novel observations about the nuclear distribution of Tcf. Furthermore, with the use of red (HcRed-AR and HcRed-Tcf) and green fusion proteins (ß-catenin-EGFP), we provide morphological evidence of a reciprocal balance of nuclear ß-catenin-EGFP (BC-EGFP). By cotransfecting in LNCaP prostate tumor cells and using quantitative imaging software, we demonstrated a 62.0% colocalization of HcRed-AR and BC-EGFP in the presence of DHT and 63.3% colocalization of HcRed-Tcf/BC-EGFP in the absence of DHT. Costaining for activated RNA Pol II (phosphoserine 2) and HcRed-Tcf suggested that Tcf foci contain transcriptional 'hotspots' validating that these sites have the capacity for transcriptional activity. Given this apparent androgen-dependent competition for nuclear BC-EGFP, we chose to assess our hypothesis by in vivo and in vitro binding assays. SW480 cells transiently transfected with an AR expression construct, treated with DHT and immunoprecipitated for Tcf showed less associated ß-catenin when compared to Tcf precipitates from untreated cells. Furthermore, by treating cells with DHT+Casodex, we were able to abrogate the androgen-sensitive AR/ß-catenin interaction, in addition to relieving transcriptional repression of the TOPFLASH reporter. In vitro binding assays, with increasing amounts of ARS35 , resulted in decreased TcfS35 association with immunoprecipitated recombinant ß-catenin-HIS. These data suggest that in steady-state conditions, AR has the ability to compete out Tcf binding for ß-catenin. Finally, using SW480 cells, we show that AR-mediated repression of the BCT pathway has implications for cell cycle progression and in vitro growth. Using FACs analysis, we observed a 26.1% increase in accumulation of cells in the G1?phase of the cell cycle, while in vitro growth assays showed a 35% reduction in viable cells transfected with AR+DHT treatment. Together, our data strongly suggest that a reciprocal balance of nuclear ß-catenin facilitates AR-mediated repression of BCT-driven transcription and cell growth.Oncogene (2003) 22, 5602-5613. doi:10.1038/sj.onc.1206802 [ABSTRACT FROM AUTHOR]- Published
- 2003
- Full Text
- View/download PDF
29. A mutant P53 can activate apoptosis through a mechanism distinct from those induced by wild type P53
- Author
-
He, Ming, Rennie, Paul S., Dragowska, Visia, Nelson, Colleen C., and Jia, William
- Subjects
GENETIC mutation ,APOPTOSIS ,PROSTATE cancer - Abstract
A common mutation in P53 protein occurs at amino acid residue 281 in the DNA binding domain (P53
gly(281) ), which results in loss of transcriptional regulation of P53 target genes and has been reported to gain pro-oncogenic functions. In the present study, we investigated the activity of P53gly(281) in P53-null PC3 human prostate cancer cells and found that the P53gly(281) induced apoptosis as efficiently as the wild-type P53 (wtP53). However, in contrast to wtP53-induced apoptosis, the P53gly(281) -induced apoptosis was insensitive to overexpression of bcl-2. Thus, our findings indicate that while a mutation in the DNA binding domain of p53 may result in a more oncogenic form of the protein, it may also paradoxically result in the ‘gain’ of a new, alternative pathway for apoptosis. [Copyright &y& Elsevier]- Published
- 2002
- Full Text
- View/download PDF
30. Engineering osteoblastic metastases to delineate the adaptive response of androgen-deprived prostate cancer in the bone metastatic microenvironment.
- Author
-
Bock, Nathalie, Shokoohmand, Ali, Kryza, Thomas, Röhl, Joan, Meijer, Jonelle, Tran, Phong A., Nelson, Colleen C., Clements, Judith A., and Hutmacher, Dietmar W.
- Subjects
OSTEOBLASTS ,PROSTATE cancer ,BONE metastasis ,ANDROGENS ,CANCER cells - Abstract
While stromal interactions are essential in cancer adaptation to hormonal therapies, the effects of bone stroma and androgen deprivation on cancer progression in bone are poorly understood. Here, we tissue-engineered and validated an in vitro microtissue model of osteoblastic bone metastases, and used it to study the effects of androgen deprivation in this microenvironment. The model was established by culturing primary human osteoprogenitor cells on melt electrowritten polymer scaffolds, leading to a mineralized osteoblast-derived microtissue containing, in a 3D setting, viable osteoblastic cells, osteocytic cells, and appropriate expression of osteoblast/osteocyte-derived mRNA and proteins, and mineral content. Direct co-culture of androgen receptor-dependent/independent cell lines (LNCaP, C4-2B, and PC3) led cancer cells to display functional and molecular features as observed in vivo. Co-cultured cancer cells showed increased affinity to the microtissues, as a function of their bone metastatic potential. Co-cultures led to alkaline phosphatase and collagen-I upregulation and sclerostin downregulation, consistent with the clinical marker profile of osteoblastic bone metastases. LNCaP showed a significant adaptive response under androgen deprivation in the microtissues, with the notable appearance of neuroendocrine transdifferentiation features and increased expression of related markers (dopa decarboxylase, enolase 2). Androgen deprivation affected the biology of the metastatic microenvironment with stronger upregulation of androgen receptor, alkaline phosphatase, and dopa decarboxylase, as seen in the transition towards resistance. The unique microtissues engineered here represent a substantial asset to determine the involvement of the human bone microenvironment in prostate cancer progression and response to a therapeutic context in this microenvironment. Cancer: 3D model mimics prostate cancer metastasis to bone A three-dimensional model of prostate tumor cells growing in a bone-like microenvironment offers a new platform for studying how metastatic prostate cancers respond to therapies. Dietmar Hutmacher and colleagues from the Queensland University of Technology in Brisbane, Australia, engineered a bone-like tissue environment by culturing bone progenitor cells in a fibrous polyester scaffold. The cells differentiated into bone-forming cells that produced the appropriate RNAs, proteins and minerals. The researchers then added various populations of prostate cancer cells to the constructs. Cell lines with the greatest bone metastatic potential grew best, and depriving cells of the hormone androgen led to a more aggressive disease phenotype consistent with that observed in the tumors of men with castration-resistant prostate cancer. The system offers a new way to test for relevant biomarkers and therapeutics in the laboratory. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
31. Androgen influence on prostate cancer exosomes production and content
- Author
-
Carolina Soekmadji, Russell, Pamela J., Hill, Michelle M., Dunne, Jonathan C., William Jordan, T., and Nelson, Colleen C.
- Subjects
biomarkers ,androgens ,exosomes ,prostate cancer ,060100 BIOCHEMISTRY AND CELL BIOLOGY
32. 572: Differential Regulation of IGFBP-3 by the Androgen Receptor in Progression to Androgen Independent Prostate Cancer.
- Author
-
Kojima, Satoko, Mulholland, David J., Nelson, Colleen C., and Gleave, Martin E.
- Subjects
ANDROGEN receptors ,ANDROGENS ,PROSTATE cancer ,PROSTATE ,DELEGATED legislation - Published
- 2005
- Full Text
- View/download PDF
33. In vitro engineering of a bone metastases model allows for study of the effects of antiandrogen therapies in advanced prostate cancer.
- Author
-
Bock, Nathalie, Kryza, Thomas, Shokoohmand, Ali, Röhl, Joan, Ravichandran, Akhilandeshwari, Wille, Marie-Luise, Nelson, Colleen C., Hutmacher, Dietmar W., and Clements, Judith A.
- Subjects
- *
ANDROGEN receptors , *DOCETAXEL , *BONE metastasis , *TREATMENT effectiveness , *PROSTATE cancer , *MEDICAL sciences , *STROMAL cell-derived factor 1 - Abstract
The article explores the in vitro engineering of a bone metastases model to study the effects of antiandrogen therapies in advanced prostate cancer. Topics include the development of an in vitro model for studying prostate cancer metastases, the effects of antiandrogen therapies (bicalutamide and enzalutamide) in a bone microenvironment, and the significance of the findings for understanding and treating metastatic prostate cancer.
- Published
- 2021
- Full Text
- View/download PDF
34. hSSB1 (NABP2/OBFC2B) modulates the DNA damage and androgen-induced transcriptional response in prostate cancer
- Author
-
Mark N. Adams, Laura V. Croft, Aaron Urquhart, Mohamed Ashick Mohamed Saleem, Anja Rockstroh, Pascal H. G. Duijf, Patrick B. Thomas, Genevieve P. Ferguson, Idris Mohd Najib, Esha T. Shah, Emma Bolderson, Shivashankar Nagaraj, Elizabeth D. Williams, Colleen C. Nelson, Kenneth J. O'Byrne, Derek J. Richard, Adams, Mark N, Croft, Laura V, Urquhart, Aaron, Saleem, Mohamed Ashick Mohamed, Rockstroh, Anja, Duijf, Pascal HG, Thomas, Patrick B, Ferguson, Genevieve P, Najib, Idris Mohd, Shah, Esha T, Bolderson, Emma, Nagaraj, Shivashankar, Williams, Elizabeth D, Nelson, Colleen C, O'Byrne, Kenneth J, and Richard, Derek J
- Subjects
radiation ,Oncology ,Urology ,androgen receptor ,DNA damage ,hSSB1/NABP2 ,prostate cancer - Abstract
Background: Activation and regulation of androgen receptor (AR) signaling and the DNA damage response impact the prostate cancer (PCa) treatment modalities of androgen deprivation therapy (ADT) and radiotherapy. Here, we have evaluated a role for human single-strand binding protein 1 (hSSB1/NABP2) in modulation of the cellular response to androgens and ionizing radiation (IR). hSSB1 has defined roles in transcription and maintenance of genome stability, yet little is known about this protein in PCa. Methods: We correlated hSSB1 with measures of genomic instability across available PCa cases from The Cancer Genome Atlas (TCGA). Microarray and subsequent pathway and transcription factor enrichment analysis were performed on LNCaP and DU145 prostate cancer cells. Results: Our data demonstrate that hSSB1 expression in PCa correlates with measures of genomic instability including multigene signatures and genomic scars that are reflective of defects in the repair of DNA double-strand breaks via homologous recombination. In response to IR-induced DNA damage, we demonstrate that hSSB1 regulates cellular pathways that control cell cycle progression and the associated checkpoints. In keeping with a role for hSSB1 in transcription, our analysis revealed that hSSB1 negatively modulates p53 and RNA polymerase II transcription in PCa. Of relevance to PCa pathology, our findings highlight a transcriptional role for hSSB1 in regulating the androgen response. We identified that AR function is predicted to be impacted by hSSB1 depletion, whereby this protein is required to modulate AR gene activity in PCa. Conclusions: Our findings point to a key role for hSSB1 in mediating the cellular response to androgen and DNA damage via modulation of transcription. Exploiting hSSB1 in PCa might yield benefits as a strategy to ensure a durable response to ADT and/or radiotherapy and improved patient outcomes. Refereed/Peer-reviewed
- Published
- 2023
35. Folate-targeted amphiphilic cyclodextrin nanoparticles incorporating a fusogenic peptide deliver therapeutic siRNA and inhibit the invasive capacity of 3D prostate cancer tumours.
- Author
-
Evans, James C., Malhotra, Meenakshi, Sweeney, Katrina, Darcy, Raphael, Nelson, Colleen C., Hollier, Brett G., and O’Driscoll, Caitriona M.
- Subjects
- *
TARGETED drug delivery , *CYCLODEXTRINS in pharmaceutical technology , *PROSTATE cancer treatment , *RNA interference , *SMALL interfering RNA - Abstract
The main barrier to the development of an effective RNA interference (RNAi) therapy is the lack of a suitable delivery vector. Modified cyclodextrins have emerged in recent years for the delivery of siRNA. In the present study, a folate-targeted amphiphilic cyclodextrin was formulated using DSPE-PEG 5000 -folate to target prostate cancer cells. The fusogenic peptide GALA was included in the formulation to aid in the endosomal release of siRNA. Targeted nanoparticles were less than 200 nm in size with a neutral surface charge. The complexes were able to bind siRNA and protect it from serum nucleases. Incubation with excess free folate resulted in a significant decrease in the uptake of targeted nanoparticles in LNCaP and PC3 cells, both of which have been reported to have differing pathways of folate uptake. There was a significant reduction in the therapeutic targets, ZEB1 and NRP1 at mRNA and protein level following treatment with targeted complexes. In preliminary functional assays using 3D spheroids, treatment of PC3 tumours with targeted complexes with ZEB1 and NRP1 siRNA resulted in more compact colonies relative to the untargeted controls and inhibited infiltration into the Matrigel™ layer. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
36. Whole-Genome Sequence of the Metastatic PC3 and LNCaP Human Prostate Cancer Cell Lines.
- Author
-
Seim, Inge, Jeffery, Penny L., Thomas, Patrick B., Nelson, Colleen C., and Chopin, Lisa K.
- Subjects
- *
NUCLEOTIDE sequencing , *PROSTATE cancer & genetics , *CELL lines - Abstract
The bone metastasis-derived PC3 and the lymph node metastasis-derived LNCaP prostate cancer cell lines are widely studied, having been described in thousands of publications over the last four decades. Here, we report short-read whole-genome sequencing (WGS) and de novo assembly of PC3 (ATCC CRL-1435) and LNCaP (clone FGC; ATCC CRL-1740) at 70 · coverage. A known homozygous mutation in TP53 and homozygous loss of PTEN were robustly identified in the PC3 cell line, whereas the LNCaP cell line exhibited a larger number of putative inactivating somatic point and indel mutations (and in particular a loss of stop codon events). This study also provides preliminary evidence that loss of one or both copies of the tumor suppressor Capicua (CIC) contributes to primary tumor relapse and metastatic progression, potentially offering a treatment target for castration-resistant prostate cancer (CRPC). Our work provides a resource for genetic, genomic, and biological studies employing two commonly-used prostate cancer cell lines. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
37. Identification of a novel fusion transcript between human relaxin-1 (RLN1) and human relaxin-2 (RLN2) in prostate cancer.
- Author
-
Tevz, Gregor, McGrath, Sean, Demeter, Ryan, Magrini, Vincent, Jeet, Varinder, Rockstroh, Anja, McPherson, Stephen, Lai, John, Bartonicek, Nenad, An, Jiyuan, Batra, Jyotsna, Dinger, Marcel E., Lehman, Melanie L., Williams, Elizabeth D., and Nelson, Colleen C.
- Subjects
- *
RELAXIN , *PROSTATE cancer , *PEPTIDES , *PROTEINS , *CORPUS luteum - Abstract
Simultaneous expression of highly homologous RLN1 and RLN2 genes in prostate impairs their accurate delineation. We used PacBio SMRT sequencing and RNA-Seq in LNCaP cells in order to dissect the expression of RLN1 and RLN2 variants. We identified a novel fusion transcript comprising the RLN1 and RLN2 genes and found evidence of its expression in the normal and prostate cancer tissues. The RLN1-RLN2 fusion putatively encodes RLN2 isoform with the deleted secretory signal peptide. The identification of the fusion transcript provided information to determine unique RLN1-RLN2 fusion and RLN1 regions. The RLN1-RLN2 fusion was co-expressed with RLN1 in LNCaP cells, but the two gene products were inversely regulated by androgens. We showed that RLN1 is underrepresented in common PCa cell lines in comparison to normal and PCa tissue. The current study brings a highly relevant update to the relaxin field, and will encourage further studies of RLN1 and RLN2 in PCa and broader. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
38. Isolation, structure determination and cytotoxicity studies of tryptophan alkaloids from an Australian marine sponge Hyrtios sp.
- Author
-
Khokhar, Shahan, Feng, Yunjiang, Campitelli, Marc R., Ekins, Merrick G., Hooper, John N.A., Beattie, Karren D., Sadowski, Martin C., Nelson, Colleen C., and Davis, Rohan A.
- Subjects
- *
TRYPTOPHAN , *SPONGES (Invertebrates) , *CELL-mediated cytotoxicity , *DENSITY functional theory , *PROSTATE cancer , *CANCER cells , *NUCLEAR magnetic resonance spectroscopy - Abstract
Mass-guided fractionation of the MeOH extract from a specimen of the Australian marine sponge Hyrtios sp. resulted in the isolation of two new tryptophan alkaloids, 6-oxofascaplysin ( 2 ), and secofascaplysic acid ( 3 ), in addition to the known metabolites fascaplysin ( 1 ) and reticulatate ( 4 ). The structures of all molecules were determined following NMR and MS data analysis. Structural ambiguities in 2 were addressed through comparison of experimental and DFT-generated theoretical NMR spectral values. Compounds 1 – 4 were evaluated for their cytotoxicity against a prostate cancer cell line (LNCaP) and were shown to display IC 50 values ranging from 0.54 to 44.9 μM. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
39. Paracrine interactions between LNCaP prostate cancer cells and bioengineered bone in 3D in vitro culture reflect molecular changes during bone metastasis.
- Author
-
Sieh, Shirly, Taubenberger, Anna V., Lehman, Melanie L., Clements, Judith A., Nelson, Colleen C., and Hutmacher, Dietmar W.
- Subjects
- *
PARACRINE mechanisms , *CELL lines , *PROSTATE cancer , *BIOENGINEERING , *CELL culture , *BONE metastasis , *IN vitro studies - Abstract
Abstract: As microenvironmental factors such as three-dimensionality and cell–matrix interactions are increasingly being acknowledged by cancer biologists, more complex 3D in vitro models are being developed to study tumorigenesis and cancer progression. To better understand the pathophysiology of bone metastasis, we have established and validated a 3D indirect co-culture model to investigate the paracrine interactions between prostate cancer (PCa) cells and human osteoblasts. Co-culture of the human PCa, LNCaP cells embedded within polyethylene glycol hydrogels with human osteoblasts in the form of a tissue engineered bone construct (TEB), resulted in reduced proliferation of LNCaP cells. LNCaP cells in both monoculture and co-culture were responsive to the androgen analog, R1881, as indicated by an increase in the expression (mRNA and/or protein induction) of androgen-regulated genes including prostate specific antigen and fatty acid synthase. Microarray gene expression analysis further revealed an up-regulation of bone markers and other genes associated with skeletal and vasculature development and a significant activation of transforming growth factor β1 downstream genes in LNCaP cells after co-culture with TEB. LNCaP cells co-cultured with TEB also unexpectedly showed similar changes in classical androgen-responsive genes under androgen-deprived conditions not seen in LNCaP monocultures. The molecular changes of LNCaP cells after co-culturing with TEBs suggest that osteoblasts exert a paracrine effect that may promote osteomimicry and modulate the expression of androgen-responsive genes in LNCaP cells. Taken together, we have presented a novel 3D in vitro model that allows the study of cellular and molecular changes occurring in PCa cells and osteoblasts that are relevant to metastatic colonization of bone. This unique in vitro model could also facilitate cancer biologists to dissect specific biological hypotheses via extensive genomic or proteomic assessments to further our understanding of the PCa-bone crosstalk. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
40. Cloning of a novel insulin-regulated ghrelin transcript in prostate cancer.
- Author
-
Seim, Inge, Lubik, Amy A., Lehman, Melanie L., Tomlinson, Nadine, Whiteside, Eliza J., Herington, Adrian C., Nelson, Colleen C., and Chopin, Lisa K.
- Subjects
- *
PROSTATE cancer , *GHRELIN , *EXONS (Genetics) , *CELL lines , *GENETIC engineering - Abstract
Ghrelin is a multifunctional hormone, with roles in stimulating appetite and regulating energy balance, insulin secretion and glucose homoeostasis. The ghrelin gene locus (GHRL) is highly complex and gives rise to a range of novel transcripts derived from alternative first exons and internally spliced exons. The wild-type transcript encodes a 117 amino acid preprohormone that is processed to yield the 28 amino acid peptide ghrelin. Here, we identified insulin-responsive transcription corresponding to cryptic exons in intron 2 of the human ghrelin gene. A transcript, termed in2c-ghrelin (intron 2-cryptic), was cloned from the testis and the LNCaP prostate cancer cell line. This transcript may encode an 83 amino acid preproghrelin isoform that codes for ghrelin, but not obestatin. It is expressed in a limited number of normal tissues and in tumours of the prostate, testis, breast and ovary. Finally, we confirmed that in2c-ghrelin transcript expression, as well as the recently described in1-ghrelin transcript, is significantly upregulated by insulin in cultured prostate cancer cells. Metabolic syndrome and hyperinsulinaemia have been associated with prostate cancer risk and progression. This may be particularly significant after androgen deprivation therapy for prostate cancer, which induces hyperinsulinaemia, and this could contribute to castrate-resistant prostate cancer growth. We have previously demonstrated that ghrelin stimulates prostate cancer cell line proliferation in vitro. This study is the first description of insulin regulation of a ghrelin transcript in cancer and should provide further impetus for studies into the expression, regulation and function of ghrelin gene products. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
41. Androgen Receptor and Nutrient Signaling Pathways Coordinate the Demand for Increased Amino Acid Transport during Prostate Cancer Progression.
- Author
-
Qian Wang, Bailey, Charles G., Ng, Cynthia, Tiffen, Jessamy, Thoeng, Annora, Minhas, Vineet, Lehman, Melanie L., Hendy, Stephen C., Buchanan, Grant, Nelson, Colleen C., Rasko, John E. J., and Holst, Jeff
- Subjects
- *
ANDROGEN receptors , *AMINO acid transport , *PROSTATE cancer , *LEUCINE , *CELL growth - Abstract
L-Type amino acid transporters such as LAT1 and LAT3 mediate the uptake of essential amino acids. Here, we report that prostate cancer cells coordinate the expression of LAT1 and LAT3 to maintain sufficient levels of leucine needed for mTORC1 signaling and cell growth. Inhibiting LAT function was sufficient to decrease cell growth and mTORC1 signaling in prostate cancer cells. These cells maintained levels of amino acid influx through androgen receptor-mediated regulation of LAT3 expression and ATF4 regulation of LAT1 expression after amino acid deprivation. These responses remained intact in primary prostate cancer, as indicated by high levels of LAT3 in primary disease, and by increased levels of LAT1 after hormone ablation and in metastatic lesions. Taken together, our results show how prostate cancer cells respond to demands for increased essential amino acids by coordinately activating amino acid transporter pathways vital for tumor outgrowth. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
42. Insulin Increases De Novo Steroidogenesis in Prostate Cancer Cells.
- Author
-
Lubik, Amy A., Gunter, Jennifer H., Hendy, Stephen C., Locke, Jennifer A., Adomat, Hans H., Thompson, Vanessa, Herington, Adrian, Gleave, Martin E., Pollak, Michael, and Nelson, Colleen C.
- Subjects
- *
ANDROGENS , *PROSTATE cancer , *METABOLIC syndrome , *INSULIN , *MESSENGER RNA - Abstract
Androgen-dependent pathways regulate maintenance and growth of normal and malignant prostate tissues. Androgen deprivation therapy (ADT) exploits this dependence and is used to treat metastatic prostate cancer; however, regression initially seen with ADT gives way to development of incurable castration-resistant prostate cancer (CRPC). Although ADT generates a therapeutic response, it is also associated with a pattern of metabolic alterations consistent with metabolic syndrome including elevated circulating insulin. Because CRPC cells are capable of synthesizing androgens de novo, we hypothesized that insulin may also influence steroidogenesis in CRPC. In this study, we examined this hypothesis by evaluating the effect of insulin on steroid synthesis in prostate cancer cell lines. Treatment with 10 nmol/L insulin increased mRNA and protein expression of steroidogenesis enzymes and upregulated the insulin receptor substrate insulin receptor substrate 2 (IRS-2). Similarly, insulin treatment upregulated intracellular testosterone levels and secreted androgens, with the concentrations of steroids observed similar to the levels reported in prostate cancer patients. With similar potency to dihydrotestosterone, insulin treatment resulted in increased mRNA expression of prostate-specific antigen. CRPC progression also correlated with increased expression of IRS-2 and insulin receptor in vivo. Taken together, our findings support the hypothesis that the elevated insulin levels associated with therapeutic castration may exacerbate progression of prostate cancer to incurable CRPC in part by enhancing steroidogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.