Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
2.
Sci Rep ; 9(1): 5120, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30914656

ABSTRACT

Loss of latexin (LXN) expression negatively correlates with the prognosis of several human cancers. Despite association with numerous processes including haematopoietic stem cell (HSC) fate, inflammation and tumour suppression, a clearly defined biological role for LXN is still lacking. Therefore, we sought to understand LXN expression and function in the normal and malignant prostate to assess its potential as a therapeutic target. Our data demonstrate that LXN is highly expressed in normal prostate luminal cells but downregulated in high Gleason grade cancers. LXN protein is both cytosolic and secreted by prostate cells and expression is directly and potently upregulated by all-trans retinoic acid (atRA). Whilst overexpression of LXN in prostate epithelial basal cells did not affect cell fate, LXN overexpression in the luminal cancer line LNCaP reduced plating efficiency. Transcriptome analysis revealed that LXN overexpression had no direct effects on gene expression but had significant indirect effects on important genes involved in both retinoid metabolism and IFN-associated inflammatory responses. These data highlight a potential role for LXN in retinoid signaling and inflammatory pathways. Investigating the effects of LXN on immune cell function in the tumour microenvironment (TME) may reveal how observed intratumoural loss of LXN affects the prognosis of many adenocarcinomas.


Subject(s)
Down-Regulation , Gene Expression Regulation, Neoplastic , Nerve Tissue Proteins/biosynthesis , Prostate/metabolism , Prostatic Neoplasms/metabolism , Tumor Suppressor Proteins/biosynthesis , Humans , Male , Nerve Tissue Proteins/genetics , PC-3 Cells , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Tumor Suppressor Proteins/genetics
3.
Nucleic Acids Res ; 47(8): 3937-3956, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30820548

ABSTRACT

RNA polymerase (pol) III occurs in two forms, containing either the POLR3G subunit or the related paralogue POLR3GL. Whereas POLR3GL is ubiquitous, POLR3G is enriched in undifferentiated cells. Depletion of POLR3G selectively triggers proliferative arrest and differentiation of prostate cancer cells, responses not elicited when POLR3GL is depleted. A small molecule pol III inhibitor can cause POLR3G depletion, induce similar differentiation and suppress proliferation and viability of cancer cells. This response involves control of the fate-determining factor NANOG by small RNAs derived from Alu short interspersed nuclear elements. Tumour initiating activity in vivo can be reduced by transient exposure to the pol III inhibitor. Untransformed prostate cells appear less sensitive than cancer cells to pol III depletion or inhibition, raising the possibility of a therapeutic window.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/drug effects , Prostatic Neoplasms/drug therapy , RNA Polymerase III/genetics , Small Molecule Libraries/pharmacology , Aged , Alu Elements/drug effects , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Endoplasmic Reticulum Chaperone BiP , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Male , Mice , Mice, Knockout , Middle Aged , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Polymerase III/antagonists & inhibitors , RNA Polymerase III/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Xenograft Model Antitumor Assays
4.
PeerJ ; 6: e5981, 2018.
Article in English | MEDLINE | ID: mdl-30498642

ABSTRACT

Patient-derived xenograft (PDX) models are increasingly being used in oncology drug development because they offer greater predictive value than traditional cell line models. Using novel tools to critique model validity and reliability we performed a systematic review to identify all original publications describing the derivation of PDX models of colon, prostate, breast and lung cancer. Validity was defined as the ability to recapitulate the disease of interest. The study protocol was registered with the Collaborative Approach to Meta-Analysis and Review of Animal Data from Experimental Studies (CAMARADES). Searches were performed in Embase, MEDLINE and Pubmed up to July 2017. A narrative data synthesis was performed. We identified 105 studies of model validations; 29 for breast, 29 for colon, 25 for lung, 23 for prostate and 4 for multiple tissues. 133 studies were excluded because they did not perform any validation experiments despite deriving a PDX. Only one study reported following the ARRIVE guidelines; developed to improve the standard of reporting for animal experimentation. Remarkably, half of all breast (52%) and prostate (50%) studies were judged to have high concern, in contrast to 16% of colon and 28% of lung studies. The validation criteria that most commonly failed (evidence to the contrary) were: tissue of origin not proven and histology of the xenograft not comparable to the parental tumour. Overall, most studies were categorized as unclear because one or more validation conditions were not reported, or researchers failed to provide data for a proportion of their models. For example, failure to demonstrate tissue of origin, response to standard of care agents and to exclude development of lymphoma. Validation tools have the potential to improve reproducibility, reduce waste in research and increase the success of translational studies.

5.
Br J Cancer ; 119(9): 1133-1143, 2018 10.
Article in English | MEDLINE | ID: mdl-30318509

ABSTRACT

BACKGROUND: Human prostate cancers display numerous DNA methylation changes compared to normal tissue samples. However, definitive identification of features related to the cells' malignant status has been compromised by the predominance of cells with luminal features in prostate cancers. METHODS: We generated genome-wide DNA methylation profiles of cell subpopulations with basal or luminal features isolated from matched prostate cancer and normal tissue samples. RESULTS: Many frequent DNA methylation changes previously attributed to prostate cancers are here identified as differences between luminal and basal cells in both normal and cancer samples. We also identified changes unique to each of the two cancer subpopulations. Those specific to cancer luminal cells were associated with regulation of metabolic processes, cell proliferation and epithelial development. Within the prostate cancer TCGA dataset, these changes were able to distinguish not only cancers from normal samples, but also organ-confined cancers from those with extraprostatic extensions. Using changes present in both basal and luminal cancer cells, we derived a new 17-CpG prostate cancer signature with high predictive power in the TCGA dataset. CONCLUSIONS: This study demonstrates the importance of comparing phenotypically matched prostate cell populations from normal and cancer tissues to unmask biologically and clinically relevant DNA methylation changes.


Subject(s)
DNA Methylation , Phenotype , Prostatic Neoplasms/genetics , CpG Islands , Humans , Male
6.
Methods Mol Biol ; 1786: 55-66, 2018.
Article in English | MEDLINE | ID: mdl-29786786

ABSTRACT

This chapter focuses on primary cultures of the human malignant prostate. Current abilities to isolate and culture stem cells, transit-amplifying cells, and secretory luminal cells are described. Advantages and limitations of this model system are also discussed.


Subject(s)
Cell Culture Techniques , Prostatic Neoplasms/pathology , Biopsy , Epithelial Cells/metabolism , Feeder Cells , Humans , Male , Spheroids, Cellular , Stromal Cells/metabolism , Tumor Cells, Cultured
7.
PLoS One ; 12(11): e0188228, 2017.
Article in English | MEDLINE | ID: mdl-29145505

ABSTRACT

Prostate cancer research is hampered by the lack of in vivo preclinical models that accurately reflect patient tumour biology and the clinical heterogeneity of human prostate cancer. To overcome these limitations we propagated and characterised a new collection of patient-derived prostate cancer xenografts. Tumour fragments from 147 unsupervised, surgical prostate samples were implanted subcutaneously into immunodeficient Rag2-/-γC-/- mice within 24 hours of surgery. Histologic and molecular characterisation of xenografts was compared with patient characteristics, including androgen-deprivation therapy, and exome sequencing. Xenografts were established from 47 of 147 (32%) implanted primary prostate cancers. Only 14% passaged successfully resulting in 20 stable lines; derived from 20 independent patient samples. Surprisingly, only three of the 20 lines (15%) were confirmed as prostate cancer; one line comprised of mouse stroma, and 16 were verified as human donor-derived lymphoid neoplasms. PCR for Epstein-Barr Virus (EBV) nuclear antigen, together with exome sequencing revealed that the lymphomas were exclusively EBV-associated. Genomic analysis determined that 14 of the 16 EBV+ lines had unique monoclonal or oligoclonal immunoglobulin heavy chain gene rearrangements, confirming their B-cell origin. We conclude that the generation of xenografts from tumour fragments can commonly result in B-cell lymphoma from patients carrying latent EBV. We recommend routine screening, of primary outgrowths, for latent EBV to avoid this phenomenon.


Subject(s)
Herpesvirus 4, Human/pathogenicity , Lymphoma/virology , Prostatic Neoplasms/virology , Aged , Heterografts , Humans , Male , Middle Aged
8.
Oncotarget ; 8(34): 56698-56713, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28915623

ABSTRACT

The PI3K/AKT/mTOR pathway is frequently activated in advanced prostate cancer, due to loss of the tumour suppressor PTEN, and is an important axis for drug development. We have assessed the molecular and functional consequences of pathway blockade by inhibiting AKT and mTOR kinases either in combination or as individual drug treatments. In established prostate cancer cell lines, a decrease in cell viability and in phospho-biomarker expression was observed. Although apoptosis was not induced, a G1 growth arrest was observed in PTEN null LNCaP cells, but not in BPH1 or PC3 cells. In contrast, when the AKT inhibitor AZD7328 was applied to patient-derived prostate cultures that retained expression of PTEN, activation of a compensatory Ras/MEK/ERK pathway was observed. Moreover, whilst autophagy was induced following treatment with AZD7328, cell viability was less affected in the patient-derived cultures than in cell lines. Surprisingly, treatment with a combination of both AZD7328 and two separate MEK1/2 inhibitors further enhanced phosphorylation of ERK1/2 in primary prostate cultures. However, it also induced irreversible growth arrest and senescence. Ex vivo treatment of a patient-derived xenograft (PDX) of prostate cancer with a combination of AZD7328 and the mTOR inhibitor KU-0063794, significantly reduced tumour frequency upon re-engraftment of tumour cells. The results demonstrate that single agent targeting of the PI3K/AKT/mTOR pathway triggers activation of the Ras/MEK/ERK compensatory pathway in near-patient samples. Therefore, blockade of one pathway is insufficient to treat prostate cancer in man.

9.
Methods Mol Biol ; 1443: 181-201, 2016.
Article in English | MEDLINE | ID: mdl-27246341

ABSTRACT

In order to fully explore the biology of a complex solid tumor such as prostate cancer, it is desirable to work with patient tissue. Only by working with cells from a tissue can we take into account patient variability and tumor heterogeneity. Cell lines have long been regarded as the workhorse of cancer research and it could be argued that they are of most use when considered within a panel of cell lines, thus taking into account specified mutations and variations in phenotype between different cell lines. However, often very different results are obtained when comparing cell lines to primary cells cultured from tissue. It stands to reason that cells cultured from patient tissue represents a close-to-patient model that should and does produce clinically relevant data. This chapter aims to illustrate the methods of processing, storing and culturing cells from prostate tissue, with a description of potential uses.


Subject(s)
Epithelial Cells/cytology , Primary Cell Culture/methods , Prostate/cytology , Prostatic Neoplasms/pathology , Stromal Cells/cytology , Tissue and Organ Harvesting/methods , Epithelial Cells/pathology , Humans , Male , Prostate/pathology , Stromal Cells/pathology , Tumor Cells, Cultured
10.
Oncotarget ; 7(32): 51965-51980, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27340920

ABSTRACT

Radiation therapy is a major primary treatment option for both localized early stage prostate cancer, and for advanced, regionally un-resectable, cancer. However, around 30% of patients still experience biochemical recurrence after radiation therapy within 10 years. Thus, identification of better biomarkers and new targets are urgently required to improve current therapeutic strategies. The miR-99 family has been shown to play an important role in the regulation of the DNA damage response, via targeting of the SWI/SNF chromatin remodeling factors, SMARCA5 and SMARCD1 in cell line models. In the present study, we have demonstrated that low expression of miR-99a and miR-100 is present in cell populations which are relatively radiation insensitive, for example in prostate cancer stem cells and in castration-resistant prostate cancer. Additionally, treatment of cells with the synthetic glucocorticoid, Dexamethasone resulted in decreased miR-99a and 100 expression, suggesting a new mechanism of miR-99a and 100 regulation in androgen-independent prostate cells. Strikingly, treatment of prostate cells with the glucocorticoid receptor inhibitor, Mifepristone was found to sensitize prostate cells to radiation by increasing the levels of miR-99a and miR-100. These results qualify the miR99 family as markers of radiation sensitivity and as potential therapeutic targets to improve efficiency of radiotherapy.


Subject(s)
MicroRNAs/metabolism , Prostatic Neoplasms/genetics , Radiation Tolerance/genetics , Receptors, Glucocorticoid/antagonists & inhibitors , Cell Line, Tumor , Dexamethasone/pharmacology , Gene Expression Regulation, Neoplastic/genetics , Glucocorticoids/pharmacology , Hormone Antagonists/pharmacology , Humans , Male , Mifepristone/pharmacology , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/pathology
11.
Eur Urol ; 69(4): 551-554, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26455356

ABSTRACT

UNLABELLED: Benign prostatic hyperplasia (BPH) treatments have changed little over many years and do not directly address the underlying cause. Because BPH is characterised by uncontrolled cell growth, the chromosomal telomeres should be eroded in the reported absence or low levels of telomerase activity, but this is not observed. We investigated the telomere biology of cell subpopulations from BPH patients undergoing transurethral resection of prostate (TURP). Measurement of TERC, TERT, and telomerase activity revealed that only the epithelial stem-like and progenitor fractions expressed high levels of telomerase activity (p<0.01) and individual enzyme components (p<0.01). Telomerase activity and TERT expression were not detected in stromal cells. Telomere length measurements reflected this activity, although the average telomere length of (telomerase-negative) luminal cells was equivalent to that of telomerase-expressing stem/progenitor cells. Immunohistochemical analysis of patient-derived BPH arrays identified distinct areas of luminal hyperproliferation, basal hyperproliferation, and basal-luminal hyperproliferation, suggesting that basal and luminal cells can proliferate independently of each other. We propose a separate lineage for the luminal and basal cell components in BPH. PATIENT SUMMARY: We unexpectedly found an enzyme called telomerase in the cells that maintain benign prostatic hyperplasia (BPH), suggesting that telomerase inhibitors could be used to alleviate BPH symptoms.


Subject(s)
Cell Lineage , Cell Proliferation , Prostate/enzymology , Prostatic Hyperplasia/enzymology , Stem Cells/enzymology , Telomerase/metabolism , Telomere Homeostasis , Biomarkers/metabolism , Humans , Male , Phenotype , Prostate/pathology , Prostate/surgery , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/surgery , RNA/metabolism , Stem Cells/pathology , Telomerase/genetics , Transurethral Resection of Prostate
12.
Br J Cancer ; 113(4): 611-5, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26203762

ABSTRACT

BACKGROUND: Objective identification of key miRNAs from transcriptomic data is difficult owing to the inherent inconsistencies within miRNA target-prediction algorithms and the promiscuous nature of miRNA-mRNA target relationship. METHODS: An integrated database of miRNAs and their 'relevant' mRNA targets was generated from validated miRNA and mRNA microarray data sets generated from patient-derived prostate epithelial normal and cancer stem-like cells (SCs) and committed basal (CB) cells. The effect of miR-542-5p inhibition was studied to provide proof-of-principle for database utility. RESULTS: Integration of miRNA-mRNA databases showed that signalling pathways and processes can be regulated by a single or relatively few miRNAs, for example, DNA repair/Notch pathway by miR-542-5p, P=0.008. Inhibition of miR-542-5p in CB cells (thereby achieving miR-542-5p expression levels similar to SCs) promoted efficient DNA repair and activated expression of Notch reporters, HES1 and Survivin, without inducing dedifferentiation into SCs. CONCLUSIONS: Our novel framework impartially identifies therapeutically relevant miRNA candidates from transcriptomic data sets.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/pathology , MicroRNAs/genetics , Prostate/metabolism , Prostate/pathology , RNA, Messenger/genetics , DNA Repair/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, Notch/genetics , Signal Transduction/genetics
13.
Eur Urol ; 67(1): 7-10, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25234358

ABSTRACT

UNLABELLED: MicroRNA (miRNA) expression profiles were generated from prostate epithelial subpopulations enriched from patient-derived benign prostatic hyperplasia (n=5), Gleason 7 treatment-naive prostate cancer (PCa) (n=5), and castration-resistant PCa (CRPC) (n=3). Microarray expression was validated in an independent patient cohort (n=10). Principal component analysis showed that miRNA expression is clustered by epithelial cell phenotype, regardless of pathologic status. We also discovered concordance between the miRNA expression profiles of unfractionated epithelial cells from CRPCs, human embryonic stem cells (SCs), and prostate epithelial SCs (both benign and malignant). MiR-548c-3p was chosen as a candidate miRNA from this group to explore its usefulness as a CRPC biomarker and/or therapeutic target. Overexpression of miR-548c-3p was confirmed in SCs (fivefold, p<0.05) and in unfractionated CRPCs (1.8-fold, p<0.05). Enforced overexpression of miR-548c-3p in differentiated cells induced stemlike properties (p<0.01) and radioresistance (p<0.01). Reanalyses of published studies further revealed that miR-548c-3p is significantly overexpressed in CRPC (p<0.05) and is associated with poor recurrence-free survival (p<0.05), suggesting that miR-548c-3p is a functional biomarker for PCa aggressiveness. Our results validate the prognostic and therapeutic relevance of miRNAs for PCa management while demonstrating that resolving cell-type and differentiation-specific differences is essential to obtain clinically relevant miRNA expression profiles. PATIENT SUMMARY: We report microRNA (miRNA) expression profiles of epithelial cell fractions from the human prostate, including stem cells. miR-548c-3p was revealed as a functional biomarker for prostate cancer progression. The evaluation of miR-548c-3p in a larger patient cohort should yield information on its clinical usefulness.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , MicroRNAs/genetics , Neoplastic Stem Cells , Prostatic Hyperplasia/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Disease-Free Survival , Epithelial Cells , Humans , Male , Oligonucleotide Array Sequence Analysis , Phenotype , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , Radiation Tolerance/genetics , Up-Regulation
14.
Stem Cell Reports ; 2(2): 180-8, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24527392

ABSTRACT

Human epithelia are organized in a hierarchical structure, where stem cells generate terminally differentiated cells via intermediate progenitors. This two-step differentiation process is conserved in all tissues, but it is not known whether a common gene set contributes to its regulation. Here, we show that retinoic acid (RA) regulates early human prostate epithelial differentiation by activating a tightly coexpressed set of 80 genes (e.g., TMPRSS2). Response kinetics suggested that some of these genes could be direct RA targets, whereas others are probably responding indirectly to RA stimulation. Comparative bioinformatic analyses of published tissue-specific microarrays and a large-scale transcriptomic data set revealed that these 80 genes are not only RA responsive but also significantly coexpressed in many human cell systems. The same gene set preferentially responds to androgens during terminal prostate epithelial differentiation, implying a cell-type-dependent interplay between RA and tissue-specific transcription factor-mediated signaling in regulating the two steps of epithelial differentiation.


Subject(s)
Cell Differentiation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/metabolism , Androgens/metabolism , Androgens/pharmacology , Biomarkers , Cell Differentiation/genetics , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Regulatory Networks , Humans , Male , Organ Specificity/genetics , Prostate/cytology , Prostate/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Tretinoin/pharmacology
15.
Cancer Res ; 73(16): 5288-98, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23824741

ABSTRACT

Interleukin (IL)-6 overexpression and constitutive STAT3 activation occur in many cancers, including prostate cancer. However, their contribution to prostate stem and progenitor cells has not been explored. In this study, we show that stem-like cells from patients with prostate cancer secrete higher levels of IL-6 than their counterparts in non-neoplastic prostate. Tumor grade did not influence the levels of expression or secretion. Stem-like and progenitor cells expressed the IL-6 receptor gp80 with concomitant expression of pSTAT3. Blockade of activated STAT3, by either anti-IL-6 antibody siltuximab (CNTO 328) or LLL12, a specific pSTAT3 inhibitor, suppressed the clonogenicity of the stem-like cells in patients with high-grade disease. In a murine xenograft model used to determine the in vivo effects of pSTAT3 suppression, LLL12 treatment effectively abolished outgrowth of a patient-derived castrate-resistant tumor. Our results indicate that the most primitive cells in prostate cancer require pSTAT3 for survival, rationalizing STAT3 as a therapeutic target to treat advanced prostate cancer.


Subject(s)
Janus Kinases/antagonists & inhibitors , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Aged , Aged, 80 and over , Animals , Anthraquinones/pharmacology , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Janus Kinases/genetics , Janus Kinases/metabolism , Male , Mice , Middle Aged , Neoplastic Stem Cells/drug effects , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Sulfonamides/pharmacology
16.
Nat Protoc ; 8(5): 836-48, 2013 May.
Article in English | MEDLINE | ID: mdl-23558784

ABSTRACT

Most cases of prostate cancer are now diagnosed as moderate-grade localized disease. These tumor specimens are important tools in the discovery and translation of prostate cancer research; however, unlike more advanced tumors, they are notoriously difficult to grow in the laboratory. We developed a system for efficiently xenografting localized human prostate cancer tissue, and we adapted this protocol to study the interactions between the specific subsets of epithelial and stromal cells. Fresh prostate tissues or isolated epithelial cells are recombined with mouse seminal vesicle mesenchyme (SVM) and grafted under the renal capsule of immunodeficient mice for optimum growth and survival. Alternatively, mouse mesenchyme can be replaced with human prostate fibroblasts in order to determine their contribution to tumor progression. Grafts can be grown for several months to determine the effectiveness of novel therapeutic compounds when administered to host mice, thereby paving the way for personalizing the treatment of individual prostate cancers.


Subject(s)
Prostatic Neoplasms/pathology , Transplantation, Heterologous/methods , Xenograft Model Antitumor Assays , Animals , Cell Culture Techniques , Cell Separation/methods , Coculture Techniques , Drug Evaluation, Preclinical , Humans , Kidney/surgery , Male , Mice , Mice, SCID , Seminal Vesicles/pathology
17.
Nat Commun ; 4: 1623, 2013.
Article in English | MEDLINE | ID: mdl-23535644

ABSTRACT

While chromosomal translocations have a fundamental role in the development of several human leukaemias, their role in solid tumour development has been somewhat more controversial. Recently, it was shown that up to 80% of prostate tumours harbour at least one such gene fusion, and that the most common fusion event, between the prostate-specific TMPRSS2 gene and the ERG oncogene, is a critical, and probably early factor in prostate cancer development. Here we demonstrate the presence and expression of this significant chromosomal rearrangement in prostate cancer stem cells. Moreover, we show that in the prostate epithelial hierarchy from both normal and tumour tissues, TMPRSS2 transcription is subjected to tight monoallelic regulation, which is retained upon asymmetric division and relaxed during epithelial cell differentiation. The presence and expression of TMPRSS2/ERG in prostate stem cells would provide ERG-driven survival advantages, allowing maintenance of this mutated genotype.


Subject(s)
Alleles , Neoplastic Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Base Sequence , Blotting, Southern , DNA Methylation , DNA Primers , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction
18.
Adv Exp Med Biol ; 777: 167-84, 2013.
Article in English | MEDLINE | ID: mdl-23161082

ABSTRACT

The origin and phenotype of stem cells in human prostate cancer remains a subject of much conjecture. In this scenario, CD133 has been successfully used as a stem cell marker in both normal prostate and prostate cancer. However, cancer stem cells have been identified without the use of this marker, opening up the possibility of a CD133 negative cancer stem cell. In this chapter, we review the current literature regarding prostate cancer stem cells, with specific reference to the expression of CD133 as a stem cell marker to identify and purify stem cells in normal prostate epithelium and prostate cancer.


Subject(s)
Neoplastic Stem Cells , Prostatic Neoplasms , Biomarkers/metabolism , Humans , Male , Neoplastic Stem Cells/metabolism , Prostatic Neoplasms/metabolism
19.
Stem Cells ; 30(6): 1087-96, 2012 06.
Article in English | MEDLINE | ID: mdl-22593016

ABSTRACT

Normal prostatic epithelium is composed of basal and luminal cells. Prostate cancer can be initiated in both benign basal and luminal stem cells, but because basal cell markers are not expressed in patient tumors, the former result was unexpected. Since the cells of origin of prostate cancer are important therapeutic targets, we sought to provide further proof that basal stem cells have tumorigenic potential. Prostatic basal cells were enriched based on α2ß1integrin(hi) expression and further enriched for stem cells using CD133 in nontumorigenic BPH-1 cells. Human embryonic stem cells (hESCs) were also used as a source of normal stem cells. To test their tumorigenicity, we used two alternate stromal-based approaches; (a) recombination with human cancer-associated fibroblasts (CAFs) or (b) recombination with embryonic stroma (urogenital mesenchyme) and treated host mice with testosterone and 17ß-estradiol. Enriched α2ß1integrin(hi) basal cells from BPH-1 cells resulted in malignant tumor formation using both assays of tumorigenicity. Surprisingly, the tumorigenic potential did not reside in the CD133(+) stem cells but was consistently observed in the CD133(-) population. CAFs also failed to induce prostatic tumors from hESCs. These data confirmed that benign human basal cells include cells of origin of prostate cancer and reinforced their importance as therapeutic targets. In addition, our data suggested that the more proliferative CD133(-) basal cells are more susceptible to tumorigenesis compared to the CD133(+)-enriched stem cells. These findings challenge the current dogma that normal stem cells and cells of origin of cancer are the same cell type(s).


Subject(s)
Antigens, CD/metabolism , Glycoproteins/metabolism , Peptides/metabolism , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , AC133 Antigen , Animals , Cell Differentiation/physiology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Immunohistochemistry , Male , Mice , Mice, SCID , Prostatic Neoplasms/metabolism
20.
Mol Cell Endocrinol ; 360(1-2): 14-24, 2012 Sep 05.
Article in English | MEDLINE | ID: mdl-21802490

ABSTRACT

The prostate gland is highly dependent on androgens for its development, growth and function. Consequently, the prostatic epithelium predominantly consists of androgen-dependent luminal cells, which express the androgen receptor at high levels. In contrast, androgens are not required for the survival of the androgen-responsive, but androgen-independent, basal compartment in which stem cells reside. Basal and luminal cells are linked in a hierarchical pathway, which most probably exists as a continuum with different stages of phenotypic change. Prostate cancer is also characterised by heterogeneity, which is reflected in its response to treatment. The putative androgen receptor negative cancer stem cell (CSC) is likely to form a resistant core after most androgen-based therapies, contributing to the evolution of castration-resistant disease. The development of CSC-targeted therapies is now of crucial importance and identifying the phenotypic differences between CSCs and both their progeny will be key in this process.


Subject(s)
Androgens/physiology , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/pathology , Androgens/metabolism , Animals , Cell Transformation, Neoplastic/metabolism , Epithelium/pathology , Gene Expression Regulation, Neoplastic , Humans , Male , Molecular Targeted Therapy , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...