Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 276, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38448753

ABSTRACT

Immune checkpoint blockade has yet to produce robust anti-cancer responses for prostate cancer. Sialyltransferases have been shown across several solid tumours, including breast, melanoma, colorectal and prostate to promote immune suppression by synthesising sialoglycans, which act as ligands for Siglec receptors. We report that ST3 beta-galactoside alpha-2,3-sialyltransferase 1 (ST3Gal1) levels negatively correlate with androgen signalling in prostate tumours. We demonstrate that ST3Gal1 plays an important role in modulating tumour immune evasion through the synthesises of sialoglycans with the capacity to engage the Siglec-7 and Siglec-9 immunoreceptors preventing immune clearance of cancer cells. Here, we provide evidence of the expression of Siglec-7/9 ligands and their respective immunoreceptors in prostate tumours. These interactions can be modulated by enzalutamide and may maintain immune suppression in enzalutamide treated tumours. We conclude that the activity of ST3Gal1 is critical to prostate cancer anti-tumour immunity and provide rationale for the use of glyco-immune checkpoint targeting therapies in advanced prostate cancer.


Subject(s)
Phenylthiohydantoin , Prostatic Neoplasms , beta-Galactoside alpha-2,3-Sialyltransferase , Male , Humans , Prostatic Neoplasms/drug therapy , Benzamides/pharmacology , Nitriles , Ligands
2.
Cancers (Basel) ; 15(14)2023 Jul 13.
Article in English | MEDLINE | ID: mdl-37509260

ABSTRACT

The methyltransferase KMT5A has been proposed as an oncogene in prostate cancer and therefore represents a putative therapeutic target. To confirm this hypothesis, we have performed a microarray study on a prostate cancer cell line model of androgen independence following KMT5A knockdown in the presence of the transcriptionally active androgen receptor (AR) to understand which genes and cellular processes are regulated by KMT5A in the presence of an active AR. We observed that 301 genes were down-regulated whilst 408 were up-regulated when KMT5A expression was reduced. KEGG pathway and gene ontology analysis revealed that apoptosis and DNA damage signalling were up-regulated in response to KMT5A knockdown whilst protein folding and RNA splicing were down-regulated. Under these conditions, the top non-AR regulated gene was found to be CDC20, a key regulator of the spindle assembly checkpoint with an oncogenic role in several cancer types. Further investigation revealed that KMT5A regulates CDC20 in a methyltransferase-dependent manner to modulate histone H4K20 methylation within its promoter region and indirectly via the p53 signalling pathway. A positive correlation between KMT5A and CDC20 expression was also observed in clinical prostate cancer samples, further supporting this association. Therefore, we conclude that KMT5A is a valid therapeutic target for the treatment of prostate cancer and CDC20 could potentially be utilised as a biomarker for effective therapeutic targeting.

3.
Cancers (Basel) ; 13(4)2021 Feb 04.
Article in English | MEDLINE | ID: mdl-33557087

ABSTRACT

Identifying novel therapeutic targets for the treatment of prostate cancer (PC) remains a key area of research. With the emergence of resistance to androgen receptor (AR)-targeting therapies, other signalling pathways which crosstalk with AR signalling are important. Over recent years, evidence has accumulated for targeting the Hippo signalling pathway. Discovered in Drosophila melanogasta, the Hippo pathway plays a role in the regulation of organ size, proliferation, migration and invasion. In response to a variety of stimuli, including cell-cell contact, nutrients and stress, a kinase cascade is activated, which includes STK4/3 and LATS1/2 to inhibit the effector proteins YAP and its paralogue TAZ. Transcription by their partner transcription factors is inhibited by modulation of YAP/TAZ cellular localisation and protein turnover. Trnascriptional enhanced associate domain (TEAD) transcription factors are their classical transcriptional partner but other transcription factors, including the AR, have been shown to be modulated by YAP/TAZ. In PC, this pathway can be dysregulated by a number of mechanisms, making it attractive for therapeutic intervention. This review looks at each component of the pathway with a focus on findings from the last year and discusses what knowledge can be applied to the field of PC.

4.
Nucleic Acids Res ; 48(10): 5366-5382, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32324216

ABSTRACT

Resistance to androgen receptor (AR) targeting therapeutics in prostate cancer (PC) is a significant clinical problem. Mechanisms by which this is accomplished include AR amplification and expression of AR splice variants, demonstrating that AR remains a key therapeutic target in advanced disease. For the first time we show that IKBKE drives AR signalling in advanced PC. Significant inhibition of AR regulated gene expression was observed upon siRNA-mediated IKBKE depletion or pharmacological inhibition due to inhibited AR gene expression in multiple cell line models including a LNCaP derivative cell line resistant to the anti-androgen, enzalutamide (LNCaP-EnzR). Phenotypically, this resulted in significant inhibition of proliferation, migration and colony forming ability suggesting that targeting IKBKE could circumvent resistance to AR targeting therapies. Indeed, pharmacological inhibition in the CWR22Rv1 xenograft mouse model reduced tumour size and enhanced survival. Critically, this was validated in patient-derived explants where enzymatic inactivation of IKBKE reduced cell proliferation and AR expression. Mechanistically, we provide evidence that IKBKE regulates AR levels via Hippo pathway inhibition to reduce c-MYC levels at cis-regulatory elements within the AR gene. Thus, IKBKE is a therapeutic target in advanced PC suggesting repurposing of clinically tested IKBKE inhibitors could be beneficial to castrate resistant PC patients.


Subject(s)
I-kappa B Kinase/physiology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Androgen/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , I-kappa B Kinase/antagonists & inhibitors , Male , Mice, Nude , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic , YAP-Signaling Proteins
5.
Oncotarget ; 6(35): 37724-36, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26462181

ABSTRACT

UNLABELLED: The androgen receptor (AR) is a key transcription factor in the initiation and progression of prostate cancer (PC) and is a major therapeutic target for the treatment of advanced disease. Unfortunately, current therapies are not curative for castration resistant PC and a better understanding of AR regulation could identify novel therapeutic targets and biomarkers to aid treatment of this disease. The AR is known to be regulated by a number of post-translational modifications and we have recently identified the deubiquitinating enzyme Usp12 as a positive regulator of AR. We determined that Usp12 deubiquitinates the AR resulting in elevated receptor stability and activity. Furthermore, Usp12 silencing was shown to reduce proliferation of PC cells.Usp12 is known to require the co-factors Uaf-1 and WDR20 for catalytic activity. In this report we focus further on the role of Uaf-1 and WDR20 in Usp12 regulation and investigate if these co-factors are also required for controlling AR activity. Firstly, we confirm the presence of the Usp12/Uaf-1/WDR20 complex in PC cells and demonstrate the importance of Uaf-1 and WDR20 for Usp12 stabilisation. Consequently, we show that individual silencing of either Uaf-1 or WDR20 is sufficient to abrogate the activity of the Usp12 complex and down-regulate AR-mediated transcription via receptor destabilisation resulting in increased apoptosis and decreased colony forming ability of PC cells. Moreover, expression of both Uaf-1 and WDR20 is higher in PC tissue compared to benign controls. Overall these results highlight the potential importance of the Usp12/Uaf-1/WDR20 complex in AR regulation and PC progression. HIGHLIGHTS: • Androgen receptor is a key transcriptional regulator in prostate cancer • Usp12/Uaf-1/WDR20 complex plays a crucial role in androgen receptor stability and activity • Destabilising an individual Usp12/Uaf-1/WDR20 complex member reduces the protein levels of the whole complex and diminishes androgen receptor activity • Protein levels of all members of the Usp12/Uaf-1/WDR20 complex are significantly increased in PC.


Subject(s)
Carrier Proteins/metabolism , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Apoptosis , Blotting, Western , Carrier Proteins/genetics , Cell Proliferation , Chromatin Immunoprecipitation , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Immunoprecipitation , Male , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Processing, Post-Translational , Signal Transduction , Transcription, Genetic , Tumor Cells, Cultured , Ubiquitin-Specific Proteases/genetics
6.
Curr Protoc Mol Biol ; 111: 26.2.1-26.2.17, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26131850

ABSTRACT

This unit provides information how to use short interfering RNA (siRNA) for sequence-specific gene silencing in mammalian cells. Several methods for siRNA generation and optimization, as well as recommendations for cell transfection and transduction, are presented.


Subject(s)
Gene Silencing , RNA Interference , Animals , Cell Line , Humans , Mammals , RNA, Small Interfering/genetics , RNA, Small Interfering/isolation & purification , RNA, Small Interfering/metabolism , Transduction, Genetic/methods , Transfection/methods
7.
Nucleic Acids Res ; 43(1): 196-207, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25488809

ABSTRACT

Endocrine therapy has successfully been used to treat estrogen receptor (ER)-positive breast cancer, but this invariably fails with cancers becoming refractory to treatment. Emerging evidence has suggested that fluctuations in ER co-regulatory protein expression may facilitate resistance to therapy and be involved in breast cancer progression. To date, a small number of enzymes that control methylation status of histones have been identified as co-regulators of ER signalling. We have identified the histone H3 lysine 9 mono- and di-methyl demethylase enzyme KDM3A as a positive regulator of ER activity. Here, we demonstrate that depletion of KDM3A by RNAi abrogates the recruitment of the ER to cis-regulatory elements within target gene promoters, thereby inhibiting estrogen-induced gene expression changes. Global gene expression analysis of KDM3A-depleted cells identified gene clusters associated with cell growth. Consistent with this, we show that knockdown of KDM3A reduces ER-positive cell proliferation and demonstrate that KDM3A is required for growth in a model of endocrine therapy-resistant disease. Crucially, we show that KDM3A catalytic activity is required for both ER-target gene expression and cell growth, demonstrating that developing compounds which target demethylase enzymatic activity may be efficacious in treating both ER-positive and endocrine therapy-resistant disease.


Subject(s)
Breast Neoplasms/enzymology , Jumonji Domain-Containing Histone Demethylases/metabolism , Receptors, Estrogen/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Jumonji Domain-Containing Histone Demethylases/physiology , MCF-7 Cells , Response Elements , Signal Transduction
8.
J Biol Chem ; 288(45): 32641-32650, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24056413

ABSTRACT

The androgen receptor (AR), a member of the nuclear receptor family, is a transcription factor involved in prostate cell growth, homeostasis, and transformation. AR is a key protein in growth and development of both normal and malignant prostate, making it a common therapeutic target in prostate cancer. AR is regulated by an interplay of multiple post-translational modifications including ubiquitination. We and others have shown that the AR is ubiquitinated by a number of E3 ubiquitin ligases, including MDM2, CHIP, and NEDD4, which can result in its proteosomal degradation or enhanced transcriptional activity. As ubiquitination of AR causes a change in AR activity or stability and impacts both survival and growth of prostate cancer cells, deubiquitination of these sites has an equally important role. Hence, deubiquitinating enzymes could offer novel therapeutic targets. We performed an siRNA screen to identify deubiquitinating enzymes that regulate AR; in that screen ubiquitin-specific protease 12 (Usp12) was identified as a novel positive regulator of AR. Usp12 is a poorly characterized protein with few known functions and requires the interaction with two cofactors, Uaf-1 and WDR20, for its enzymatic activity. In this report we demonstrate that Usp12, in complex with Uaf-1 and WDR20, deubiquitinates the AR to enhance receptor stability and transcriptional activity. Our data show that Usp12 acts in a pro-proliferative manner by stabilizing AR and enhancing its cellular function.


Subject(s)
Cell Proliferation , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , COS Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Humans , Male , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Prostatic Neoplasms/genetics , Protein Stability , Receptors, Androgen/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin-Protein Ligases/genetics
9.
Nucleic Acids Res ; 41(14): 6892-904, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23723241

ABSTRACT

The importance of the estrogen receptor (ER) in breast cancer (BCa) development makes it a prominent target for therapy. Current treatments, however, have limited effectiveness, and hence the definition of new therapeutic targets is vital. The ER is a member of the nuclear hormone receptor superfamily of transcription factors that requires co-regulator proteins for complete regulation. Emerging evidence has implicated a small number of histone methyltransferase (HMT) and histone demethylase (HDM) enzymes as regulators of ER signalling, including the histone H3 lysine 9 tri-/di-methyl HDM enzyme KDM4B. Two recent independent reports have demonstrated that KDM4B is required for ER-mediated transcription and depletion of the enzyme attenuates BCa growth in vitro and in vivo. Here we show that KDM4B has an overarching regulatory role in the ER signalling cascade by controlling expression of the ER and FOXA1 genes, two critical components for maintenance of the estrogen-dependent phenotype. KDM4B interacts with the transcription factor GATA-3 in BCa cell lines and directly co-activates GATA-3 activity in reporter-based experiments. Moreover, we reveal that KDM4B recruitment and demethylation of repressive H3K9me3 marks within upstream regulatory regions of the ER gene permits binding of GATA-3 to drive receptor expression. Ultimately, our findings confirm the importance of KDM4B within the ER signalling cascade and as a potential therapeutic target for BCa treatment.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/metabolism , Receptors, Estrogen/metabolism , Signal Transduction , Cell Line , GATA3 Transcription Factor/metabolism , Gene Expression Regulation , Hepatocyte Nuclear Factor 3-alpha/metabolism , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/physiology , MCF-7 Cells , Receptors, Estrogen/genetics , Regulatory Elements, Transcriptional
10.
Nucleic Acids Res ; 41(8): 4433-46, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23435229

ABSTRACT

The androgen receptor (AR) is a key molecule involved in prostate cancer (PC) development and progression. Post-translational modification of the AR by co-regulator proteins can modulate its transcriptional activity. To identify which demethylases might be involved in AR regulation, an siRNA screen was performed to reveal that the demethylase, KDM4B, may be an important co-regulator protein. KDM4B enzymatic activity is required to enhance AR transcriptional activity; however, independently of this activity, KDM4B can enhance AR protein stability via inhibition of AR ubiquitination. Importantly, knockdown of KDM4B in multiple cell lines results in almost complete depletion of AR protein levels. For the first time, we have identified KDM4B to be an androgen-regulated demethylase enzyme, which can influence AR transcriptional activity not only via demethylation activity but also via modulation of ubiquitination. Together, these findings demonstrate the close functional relationship between AR and KDM4B, which work together to amplify the androgen response. Furthermore, KDM4B expression in clinical PC specimens positively correlates with increasing cancer grade (P < 0.001). Consequently, KDM4B is a viable therapeutic target in PC.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/metabolism , Receptors, Androgen/metabolism , Androgens/pharmacology , Animals , Cell Line , Cell Proliferation , Gene Expression Regulation , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/physiology , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Protein Processing, Post-Translational , Protein Stability , Signal Transduction , Transcription, Genetic , Ubiquitination
11.
PLoS One ; 7(11): e48944, 2012.
Article in English | MEDLINE | ID: mdl-23145034

ABSTRACT

Stem cells are thought to be the cell of origin in malignant transformation in many tissues, but their role in human prostate carcinogenesis continues to be debated. One of the conflicts with this model is that cancer stem cells have been described to lack androgen receptor (AR) expression, which is of established importance in prostate cancer initiation and progression. We re-examined the expression patterns of AR within adult prostate epithelial differentiation using an optimised sensitive and specific approach examining transcript, protein and AR regulated gene expression. Highly enriched populations were isolated consisting of stem (α(2)ß(1)(HI) CD133(+VE)), transiently amplifying (α(2)ß(1)(HI) CD133(-VE)) and terminally differentiated (α(2)ß(1)(LOW) CD133(-VE)) cells. AR transcript and protein expression was confirmed in α(2)ß(1)(HI) CD133(+VE) and CD133(-VE) progenitor cells. Flow cytometry confirmed that median (±SD) fraction of cells expressing AR were 77% (±6%) in α(2)ß(1)(HI) CD133(+VE) stem cells and 68% (±12%) in α(2)ß(1)(HI) CD133(-VE) transiently amplifying cells. However, 3-fold lower levels of total AR protein expression (peak and median immunofluorescence) were present in α(2)ß(1)(HI) CD133(+VE) stem cells compared with differentiated cells. This finding was confirmed with dual immunostaining of prostate sections for AR and CD133, which again demonstrated low levels of AR within basal CD133(+VE) cells. Activity of the AR was confirmed in prostate progenitor cells by the expression of low levels of the AR regulated genes PSA, KLK2 and TMPRSS2. The confirmation of AR expression in prostate progenitor cells allows integration of the cancer stem cell theory with the established models of prostate cancer initiation based on a functional AR. Further study of specific AR functions in prostate stem and differentiated cells may highlight novel mechanisms of prostate homeostasis and insights into tumourigenesis.


Subject(s)
Antigens, CD/metabolism , Cell Transformation, Neoplastic/metabolism , Glycoproteins/metabolism , Neoplastic Stem Cells/metabolism , Peptides/metabolism , Prostate/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Stem Cells/metabolism , AC133 Antigen , Antigens, CD/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression , Glycoproteins/genetics , Humans , Male , Neoplastic Stem Cells/pathology , Peptides/genetics , Prostate/cytology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Stem Cells/cytology
12.
PLoS One ; 7(10): e45539, 2012.
Article in English | MEDLINE | ID: mdl-23056207

ABSTRACT

Tip60 (KAT5) is a histone acetyltransferase (HAT enzyme) involved in multiple cellular processes including transcriptional regulation, DNA damage repair and cell signalling. In prostate cancer, aggressive cases over-express Tip60 which functions as an androgen receptor co-activator via direct acetylation of lysine residues within the KLKK motif of the receptor hinge region. The purpose of this study was to identify and characterise a Tip60 acetylase inhibitor. High-throughput screening revealed an isothiazole that inhibited both Tip60 and p300 HAT activity. This substance (initially identified as 4-methyl-5-bromoisothiazole) and other isothiazoles were synthesised and assayed against Tip60. Although an authentic sample of 4-methyl-5-bromoisothiazole was inactive against Tip60, in an in vitro HAT assay, 1,2-bis(isothiazol-5-yl)disulfane (NU9056) was identified as a relatively potent inhibitor (IC(50) 2 µM). Cellular activity was confirmed by analysis of acetylation of histone and non-histone proteins in a prostate cancer cell line model. NU9056 treatment inhibited cellular proliferation in a panel of prostate cancer cell lines (50% growth inhibition, 8-27 µM) and induced apoptosis via activation of caspase 3 and caspase 9 in a concentration- and time-dependent manner. Also, decreased androgen receptor, prostate specific antigen, p53 and p21 protein levels were demonstrated in response to treatment with NU9056. Furthermore, pre-treatment with NU9056 inhibited both ATM phosphorylation and Tip60 stabilization in response to ionising radiation. Based on the activity of NU9056 and the specificity of the compound towards Tip60 relative to other HAT enzymes, these chemical biology studies have identified Tip60 as a potential therapeutic target for the treatment of prostate cancer.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Histone Acetyltransferases/antagonists & inhibitors , Thiazoles/pharmacology , Acetylation/drug effects , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , DNA-Binding Proteins/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , HEK293 Cells , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histones/metabolism , Humans , Inhibitory Concentration 50 , Lysine Acetyltransferase 5 , Male , Models, Chemical , Molecular Structure , Phosphorylation/drug effects , Phosphorylation/radiation effects , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Radiation, Ionizing , Thiazoles/chemistry , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
13.
J Endocrinol ; 215(2): 221-37, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22872761

ABSTRACT

The androgen receptor (AR) is a key molecule in prostate cancer and Kennedy's disease. Understanding the regulatory mechanisms of this steroid receptor is important in the development of potential therapies for these diseases. One layer of AR regulation is provided by post-translational modifications including phosphorylation, acetylation, sumoylation, ubiquitination and methylation. While these modifications have mostly been studied as individual events, it is becoming clear that these modifications can functionally interact with each other in a signalling pathway. In this review, the effects of all modifications are described with a focus on interplay between them and the functional consequences for the AR.


Subject(s)
Protein Processing, Post-Translational/physiology , Receptors, Androgen/metabolism , Androgen Receptor Antagonists/pharmacology , Androgens/pharmacology , Animals , Protein Processing, Post-Translational/genetics , Receptors, Androgen/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...