Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Publication year range
1.
J Med Virol ; 96(3): e29540, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38529542

ABSTRACT

The sex disparity in COVID-19 outcomes with males generally faring worse than females has been associated with the androgen-regulated expression of the protease TMPRSS2 and the cell receptor ACE2 in the lung and fueled interest in antiandrogens as potential antivirals. In this study, we explored enzalutamide, an antiandrogen used commonly to treat prostate cancer, as a potential antiviral against the human coronaviruses which cause seasonal respiratory infections (HCoV-NL63, -229E, and -OC43). Using lentivirus-pseudotyped and authentic HCoV, we report that enzalutamide reduced 229E and NL63 entry and infection in both TMPRSS2- and nonexpressing immortalized cells, suggesting a TMPRSS2-independent mechanism. However, no effect was observed against OC43. To decipher this distinction, we performed RNA-sequencing analysis on 229E- and OC43-infected primary human airway cells. Our results show a significant induction of androgen-responsive genes by 229E compared to OC43 at 24 and 72 h postinfection. The virus-mediated effect on AR-signaling was further confirmed with a consensus androgen response element-driven luciferase assay in androgen-depleted MRC-5 cells. Specifically, 229E induced luciferase-reporter activity in the presence and absence of the synthetic androgen mibolerone, while OC43 inhibited induction. These findings highlight a complex interplay between viral infections and androgen-signaling, offering insights for disparities in viral outcomes and antiviral interventions.


Subject(s)
Androgens , Benzamides , Coronavirus 229E, Human , Nitriles , Phenylthiohydantoin , Male , Female , Humans , Androgens/metabolism , Androgens/pharmacology , Androgen Antagonists/pharmacology , Androgen Antagonists/metabolism , Seasons , Antiviral Agents/pharmacology , Antiviral Agents/metabolism , Luciferases
2.
Br J Cancer ; 128(12): 2326-2337, 2023 06.
Article in English | MEDLINE | ID: mdl-37076563

ABSTRACT

BACKGROUND: Current strategies to inhibit androgen receptor (AR) are circumvented in castration-resistant prostate cancer (CRPC). Cyclin-dependent kinase 7 (CDK7) promotes AR signalling, in addition to established roles in cell cycle and global transcription, providing a rationale for its therapeutic targeting in CRPC. METHODS: The antitumour activity of CT7001, an orally bioavailable CDK7 inhibitor, was investigated across CRPC models in vitro and in xenograft models in vivo. Cell-based assays and transcriptomic analyses of treated xenografts were employed to investigate the mechanisms driving CT7001 activity, alone and in combination with the antiandrogen enzalutamide. RESULTS: CT7001 selectively engages with CDK7 in prostate cancer cells, causing inhibition of proliferation and cell cycle arrest. Activation of p53, induction of apoptosis, and suppression of transcription mediated by full-length and constitutively active AR splice variants contribute to antitumour efficacy in vitro. Oral administration of CT7001 represses growth of CRPC xenografts and significantly augments growth inhibition achieved by enzalutamide. Transcriptome analyses of treated xenografts indicate cell cycle and AR inhibition as the mode of action of CT7001 in vivo. CONCLUSIONS: This study supports CDK7 inhibition as a strategy to target deregulated cell proliferation and demonstrates CT7001 is a promising CRPC therapeutic, alone or in combination with AR-targeting compounds.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Male , Humans , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Cell Line, Tumor , Xenograft Model Antitumor Assays , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Nitriles/therapeutic use , Cyclin-Dependent Kinases/therapeutic use , Enzyme Inhibitors/therapeutic use , Cell Proliferation
3.
J Allergy Clin Immunol ; 149(1): 237-251.e12, 2022 01.
Article in English | MEDLINE | ID: mdl-33964300

ABSTRACT

BACKGROUND: Allergic asthma is more severe and frequent in women than in men. In male mice, androgens negatively control group 2 innate lymphoid cell (ILC2) development and function by yet unknown mechanisms. OBJECTIVES: We sought to investigate the impact of androgen on ILC2 homeostasis and IL-33-mediated inflammation in female lungs. We evaluated the role of androgen receptor (AR) signaling and the contribution of the putative inhibitory receptor killer cell lectin-like receptor G1 (KLRG1). METHODS: Subcutaneous pellets mimicking physiological levels of androgen were used to treat female mice together with mice expressing a reporter enzyme under the control of androgen response elements and mixed bone marrow chimeras to assess the cell-intrinsic role of AR activation within ILC2s. We generated KLRG1-deficient mice. RESULTS: We established that lung ILC2s express a functionally active AR that can be in vivo targeted with exogenous androgens to negatively control ILC2 homeostasis, proliferation, and function. Androgen signaling upregulated KLRG1 on ILC2s, which inhibited their proliferation on E-cadherin interaction. Despite evidence that KLRG1 impaired the competitive fitness of lung ILC2s during inflammation, KLRG1 deficiency neither alters in vivo ILC2 numbers and functions, nor did it lead to hyperactive ILC2s in either sexes. CONCLUSIONS: AR agonists can be used in vivo to inhibit ILC2 homeostatic numbers and ILC2-dependent lung inflammation through cell-intrinsic AR activation. Although androgen signals in ILC2s to upregulate KLRG1, we demonstrate that KLRG1 is dispensable for androgen-mediated inhibition of pulmonary ILC2s.


Subject(s)
Androgens/pharmacology , Lectins, C-Type/immunology , Lymphocytes/immunology , Pneumonia/immunology , Receptors, Immunologic/immunology , Testosterone/pharmacology , Animals , Female , Interleukin-33/immunology , Lung/immunology , Lung/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/pathology , Sex Characteristics , Signal Transduction
4.
Br J Cancer ; 126(3): 502-513, 2022 02.
Article in English | MEDLINE | ID: mdl-35022525

ABSTRACT

BACKGROUND: Reliable prognostic biomarkers to distinguish indolent from aggressive prostate cancer (PCa) are lacking. Many studies investigated microRNAs (miRs) as PCa prognostic biomarkers, often reporting inconsistent findings. We present a systematic review of these; also systematic reanalysis of public miR-profile datasets to identify tissue-derived miRs prognostic of biochemical recurrence (BCR) in patients undergoing radical prostatectomy. METHODS: Independent PubMed searches were performed for relevant articles from January 2007 to December 2019. For the review, 128 studies were included. Pooled-hazard-ratios (HRs) for miRs in multiple studies were calculated using a random-effects model (REM). For the reanalysis, five studies were included and Cox proportional-hazard models, testing miR association with BCR, performed for miRs profiled in all. RESULTS: Systematic review identified 120 miRs as prognostic. Five (let-7b-5p, miR-145-5p, miR152-3p, miR-195-5p, miR-224-5p) were consistently associated with progression in multiple cohorts/studies. In the reanalysis, ten (let-7a-5p, miR-148a-3p, miR-203a-3p, miR-26b-5p, miR30a-3p, miR-30c-5p, miR-30e-3p, miR-374a-5p, miR-425-3p, miR-582-5p) were significantly prognostic of BCR. Of these, miR-148a-3p (HR = 0.80/95% CI = 0.68-0.94) and miR-582-5p (HR = 0.73/95% CI = 0.61-0.87) were also reported in prior publication(s) in the review. CONCLUSIONS: Fifteen miRs were consistently associated with disease progression in multiple publications or datasets. Further research into their biological roles is warranted to support investigations into their performance as prognostic PCa biomarkers.


Subject(s)
Biomarkers, Tumor/genetics , MicroRNAs/genetics , Prostatectomy/methods , Prostatic Neoplasms/pathology , Cohort Studies , Humans , Male , Prognosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/surgery , Survival Rate
5.
FASEB J ; 35(4): e21464, 2021 04.
Article in English | MEDLINE | ID: mdl-33724574

ABSTRACT

Chemical castration in prostate cancer can be achieved with gonadotropin-releasing hormone (GnRH) agonists or antagonists. Their effects differ by the initial flare of gonadotropin and testosterone secretion with agonists and the immediate pituitary-testicular suppression by antagonists. While both suppress luteinizing hormone (LH) and follicle-stimulating hormone (FSH) initially, a rebound in FSH levels occurs during agonist treatment. This rebound is potentially harmful, taken the expression of FSH receptors (R) in prostate cancer tissue. We herein assessed the role of FSH in promoting the growth of androgen-independent (PC-3, DU145) and androgen-dependent (VCaP) human prostate cancer cell line xenografts in nude mice. Gonadotropins were suppressed with the GnRH antagonist degarelix, and effects of add-back human recombinant FSH were assessed on tumor growth. All tumors expressed GnRHR and FSHR, and degarelix treatment suppressed their growth. FSH supplementation reversed the degarelix-evoked suppression of PC-3 tumors, both in preventive (degarelix and FSH treatment started upon cell inoculation) and therapeutic (treatments initiated 3 weeks after cell inoculation) setting. A less marked, though significant FSH effect occurred in DU145, but not in VCaP xenografts. FSHR expression in the xenografts supports direct FSH stimulation of tumor growth. Testosterone supplementation, to maintain the VCaP xenografts, apparently masked the FSH effect on their growth. Treatment with the LH analogue hCG did not affect PC-3 tumor growth despite their expression of luteinizing hormone/choriongonadotropin receptor. In conclusion, FSH, but not LH, may directly stimulate the growth of androgen-independent prostate cancer, suggesting that persistent FSH suppression upon GnRH antagonist treatment offers a therapeutic advantage over agonist.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Heterografts/drug effects , Luteinizing Hormone/metabolism , Prostatic Neoplasms/drug therapy , Androgens/pharmacology , Animals , Cell Line , Follicle Stimulating Hormone/metabolism , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Humans , Male , Mice, Nude , Prostatic Neoplasms/metabolism , Receptors, FSH , Testis/metabolism , Testosterone/pharmacology
6.
Adv Exp Med Biol ; 1390: 277-293, 2022.
Article in English | MEDLINE | ID: mdl-36107325

ABSTRACT

The androgen receptor (AR) is a ligand-activated transcription factor belonging to the nuclear receptor (NR) superfamily. As with other members of the NR family, transcriptional activity of the AR is regulated by interactions with coregulatory proteins, which either enhance (coactivators) or repress (corepressors) its transcriptional activity. AR associated coregulators are functionally diverse, but a large fraction are epigenetic histone and chromatin modifiers. Epigenetic coregulators are recruited to gene regulatory regions as part of multi-protein complexes, often acting in a dynamic and inter-dependent manner to remodel chromatin, thereby allowing or inhibiting the access of AR-associated transcriptional machinery to target genes; functional consequences being regulation of transcriptional output. Epigenetic modifiers, including those that function as AR coregulators, are frequently mutated or aberrantly expressed in prostate cancer and are implicated in disease progression. Some of these modifiers are being investigated as therapeutic targets in several cancer types and could potentially be used to modulate aberrant AR activity in prostate cancer. In this chapter we will summarise the functional role of epigenetic coregulators in AR signalling, their dysregulation during prostate cancer progression and the current status of drugs targeting these enzymes.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Chromatin , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Epigenesis, Genetic , Histones/genetics , Histones/metabolism , Humans , Ligands , Male , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Transcription Factors/metabolism
7.
Proc Natl Acad Sci U S A ; 116(34): 16711-16716, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31371493

ABSTRACT

To date, reconstitution of one of the fundamental methods of cell communication, the signaling pathway, has been unaddressed in the bottom-up construction of artificial cells (ACs). Such developments are needed to increase the functionality and biomimicry of ACs, accelerating their translation and application in biotechnology. Here, we report the construction of a de novo synthetic signaling pathway in microscale nested vesicles. Vesicle-cell models respond to external calcium signals through activation of an intracellular interaction between phospholipase A2 and a mechanosensitive channel present in the internal membranes, triggering content mixing between compartments and controlling cell fluorescence. Emulsion-based approaches to AC construction are therefore shown to be ideal for the quick design and testing of new signaling networks and can readily include synthetic molecules difficult to introduce to biological cells. This work represents a foundation for the engineering of multicompartment-spanning designer pathways that can be utilized to control downstream events inside an AC, leading to the assembly of micromachines capable of sensing and responding to changes in their local environment.


Subject(s)
Artificial Cells , Cell Compartmentation , Mechanotransduction, Cellular , Calcium/metabolism , Cell Communication/drug effects , Cell Compartmentation/drug effects , Chelating Agents/pharmacology , Escherichia coli Proteins/metabolism , Ion Channels/metabolism , Mechanotransduction, Cellular/drug effects , Phospholipases A2/metabolism
8.
J Am Chem Soc ; 143(23): 8911-8924, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34085829

ABSTRACT

Kallikrein-related peptidases (KLKs) are a family of secreted serine proteases, which form a network (the KLK activome) with an important role in proteolysis and signaling. In prostate cancer (PCa), increased KLK activity promotes tumor growth and metastasis through multiple biochemical pathways, and specific quantification and tracking of changes in the KLK activome could contribute to validation of KLKs as potential drug targets. Herein we report a technology platform based on novel activity-based probes (ABPs) and inhibitors enabling simultaneous orthogonal analysis of KLK2, KLK3, and KLK14 activity in hormone-responsive PCa cell lines and tumor homogenates. Importantly, we identifed a significant decoupling of KLK activity and abundance and suggest that KLK proteolysis should be considered as an additional parameter, along with the PSA blood test, for accurate PCa diagnosis and monitoring. Using selective inhibitors and multiplexed fluorescent activity-based protein profiling (ABPP), we dissect the KLK activome in PCa cells and show that increased KLK14 activity leads to a migratory phenotype. Furthermore, using biotinylated ABPs, we show that active KLK molecules are secreted into the bone microenvironment by PCa cells following stimulation by osteoblasts suggesting KLK-mediated signaling mechanisms could contribute to PCa metastasis to bone. Together our findings show that ABPP is a powerful approach to dissect dysregulation of the KLK activome as a promising and previously underappreciated therapeutic target in advanced PCa.


Subject(s)
Antineoplastic Agents/pharmacology , Coumarins/pharmacology , Enzyme Inhibitors/pharmacology , Kallikreins/antagonists & inhibitors , Prostate-Specific Antigen/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Coumarins/chemistry , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Humans , Kallikreins/metabolism , Male , Molecular Structure , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
10.
Nucleic Acids Res ; 45(5): 2809-2828, 2017 03 17.
Article in English | MEDLINE | ID: mdl-27907888

ABSTRACT

Regulation of microRNA (miR) biogenesis is complex and stringently controlled. Here, we identify the kinase GSK3ß as an important modulator of miR biogenesis at Microprocessor level. Repression of GSK3ß activity reduces Drosha activity toward pri-miRs, leading to accumulation of unprocessed pri-miRs and reduction of pre-miRs and mature miRs without altering levels or cellular localisation of miR biogenesis proteins. Conversely, GSK3ß activation increases Drosha activity and mature miR accumulation. GSK3ß achieves this through promoting Drosha:cofactor and Drosha:pri-miR interactions: it binds to DGCR8 and p72 in the Microprocessor, an effect dependent upon presence of RNA. Indeed, GSK3ß itself can immunoprecipitate pri-miRs, suggesting possible RNA-binding capacity. Kinase assays identify the mechanism for GSK3ß-enhanced Drosha activity, which requires GSK3ß nuclear localisation, as phosphorylation of Drosha at S300 and/or S302; confirmed by enhanced Drosha activity and association with cofactors, and increased abundance of mature miRs in the presence of phospho-mimic Drosha. Functional implications of GSK3ß-enhanced miR biogenesis are illustrated by increased levels of GSK3ß-upregulated miR targets following GSK3ß inhibition. These data, the first to link GSK3ß with the miR cascade in humans, highlight a novel pro-biogenesis role for GSK3ß in increasing miR biogenesis as a component of the Microprocessor complex with wide-ranging functional consequences.

11.
Int J Mol Sci ; 20(21)2019 Oct 29.
Article in English | MEDLINE | ID: mdl-31671745

ABSTRACT

Liver X receptors (LXRs) are ligand-dependent transcription factors acting as 'cholesterol sensors' to regulate lipid homeostasis in cells. The two isoforms, LXRα (NR1H3) and LXRß (NR1H2), are differentially expressed, with the former expressed predominantly in metabolically active tissues and the latter more ubiquitously. Both are activated by oxidised cholesterol metabolites, endogenously produced oxysterols. LXRs have important roles in lipid metabolism and inflammation, plus a number of newly emerging roles. They are implicated in regulating lipid balance in normal male reproductive function and may provide a link between male infertility and lipid disorders and/or obesity. Studies from Lxr knockout mouse models provide compelling evidence to support this. More recently published data suggest distinct and overlapping roles of the LXR isoforms in the testis and recent evidence of a role for LXRs in human male fertility. This review summarises the current literature and explores the likely link between LXR, lipid metabolism and male fertility as part of a special issue on Liver X receptors in International Journal of Molecular Sciences.


Subject(s)
Fertility/physiology , Infertility, Male/metabolism , Liver X Receptors/metabolism , Animals , Cholesterol/metabolism , Epididymis/growth & development , Epididymis/metabolism , Gene Expression Regulation , Homeostasis , Humans , Inflammation/metabolism , Lipid Metabolism , Lipids/physiology , Liver X Receptors/chemistry , Liver X Receptors/genetics , Male , Mice , Mice, Knockout , Obesity/metabolism , Oxysterols/metabolism , Protein Isoforms/metabolism , Sertoli Cells/metabolism , Testis/growth & development , Testis/metabolism
12.
Rapid Commun Mass Spectrom ; 31(19): 1599-1606, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28703389

ABSTRACT

RATIONALE: A novel data-independent acquisition method is detailed that incorporates a scanning quadrupole in front of an orthogonal acceleration time-of-flight (TOF) mass analyser. This approach is described and the attributes are compared and contrasted to other DIA approaches. METHODS: Specific application of the method to both targeted and untargeted lipidomic identification strategies is discussed, with data from both shotgun and LC separated lipidomics experiments presented. RESULTS: The benefits of the fast quadrupole scanning technique are highlighted, and include improvements in speed and specificity for complex mixtures providing high quality qualitative and quantitative data. CONCLUSIONS: In particular the high specificity afforded by the scanning quadrupole improves qualitative information for lipid identification.


Subject(s)
Chromatography, High Pressure Liquid/methods , Lipids/analysis , Mass Spectrometry/methods , Animals , Cattle , Male , Metabolomics/methods , Mice , Myocardium/chemistry , Testis/chemistry
13.
Mol Cell Proteomics ; 14(5): 1201-16, 2015 May.
Article in English | MEDLINE | ID: mdl-25693800

ABSTRACT

Current therapies for prostate cancer include antiandrogens, inhibitory ligands of the androgen receptor, which repress androgen-stimulated growth. These include the selective androgen receptor modulators cyproterone acetate and hydroxyflutamide and the complete antagonist bicalutamide. Their activity is partly dictated by the presence of androgen receptor mutations, which are commonly detected in patients who relapse while receiving antiandrogens, i.e. in castrate-resistant prostate cancer. To characterize the early proteomic response to these antiandrogens we used the LNCaP prostate cancer cell line, which harbors the androgen receptor mutation most commonly detected in castrate-resistant tumors (T877A), analyzing alterations in the proteome, and comparing these to the effect of these therapeutics upon androgen receptor activity and cell proliferation. The majority are regulated post-transcriptionally, possibly via nongenomic androgen receptor signaling. Differences detected between the exposure groups demonstrate subtle changes in the biological response to each specific ligand, suggesting a spectrum of agonistic and antagonistic effects dependent on the ligand used. Analysis of the crystal structures of the AR in the presence of cyproterone acetate, hydroxyflutamide, and DHT identified important differences in the orientation of key residues located in the AF-2 and BF-3 protein interaction surfaces. This further implies that although there is commonality in the growth responses between androgens and those antiandrogens that stimulate growth in the presence of a mutation, there may also be influential differences in the growth pathways stimulated by the different ligands. This therefore has implications for prostate cancer treatment because tumors may respond differently dependent upon which mutation is present and which ligand is activating growth, also for the design of selective androgen receptor modulators, which aim to elicit differential proteomic responses dependent upon cellular context.


Subject(s)
Androgen Antagonists/pharmacology , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/isolation & purification , Prostate/drug effects , Proteome/isolation & purification , Receptors, Androgen/chemistry , Amino Acid Sequence , Androgen Antagonists/chemistry , Anilides/chemistry , Anilides/pharmacology , Cell Line, Tumor , Cyproterone Acetate/chemistry , Cyproterone Acetate/pharmacology , Flutamide/analogs & derivatives , Flutamide/chemistry , Flutamide/pharmacology , Humans , Male , Molecular Sequence Annotation , Molecular Sequence Data , Mutation , Nandrolone/analogs & derivatives , Nandrolone/chemistry , Nandrolone/pharmacology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nitriles/chemistry , Nitriles/pharmacology , Prostate/metabolism , Prostate/pathology , Proteome/genetics , Proteome/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction , Tosyl Compounds/chemistry , Tosyl Compounds/pharmacology
14.
Anal Chem ; 88(16): 8091-8, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27498854

ABSTRACT

Highly abundant in cells, microRNAs (or miRs) play a key role as regulators of gene expression. A proportion of them are also detectable in biofluids making them ideal noninvasive biomarkers for pathologies in which miR levels are aberrantly expressed, such as cancer. Peptide nucleic acids (PNAs) are engineered uncharged oligonucleotide analogues capable of hybridizing to complementary nucleic acids with high affinity and high specificity. Herein, novel PNA-based fluorogenic biosensors have been designed and synthesized that target miR biomarkers for prostate cancer (PCa). The sensing strategy is based on oligonucleotide-templated reactions where the only miR of interest serves as a matrix to catalyze an otherwise highly unfavorable fluorogenic reaction. Validated in vitro using synthetic RNAs, these newly developed biosensors were then shown to detect endogenous concentrations of miR in human blood samples without the need for any amplification step and with minimal sample processing. This low-cost, quantitative, and versatile sensing technology has been technically validated using gold-standard RT-qPCR. Compared to RT-qPCR however, this enzyme-free, isothermal blood test is amenable to incorporation into low-cost portable devices and could therefore be suitable for widespread public screening.


Subject(s)
Biomarkers/blood , Biosensing Techniques , Circulating MicroRNA/blood , Fluorescent Dyes/chemistry , Nucleic Acid Probes/chemistry , Peptide Nucleic Acids/chemistry , Prostatic Neoplasms/diagnosis , Circulating MicroRNA/genetics , Circulating MicroRNA/metabolism , Coumarins/chemistry , Humans , Male , Nucleic Acid Hybridization , Nucleic Acid Probes/metabolism , Oligonucleotides/chemistry , Peptide Nucleic Acids/chemical synthesis , Polymorphism, Single Nucleotide , Prostatic Neoplasms/genetics , Real-Time Polymerase Chain Reaction
15.
Hum Mol Genet ; 21(14): 3112-27, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22505583

ABSTRACT

MicroRNAs (miRs) play an important role in the development of many complex human diseases and may have tumour suppressor or oncogenic (oncomir) properties. Prostate cancer is initially an androgen-driven disease, and androgen receptor (AR) remains a key driver of growth even in castration-resistant tumours. However, AR-mediated oncomiR pathways remain to be elucidated. We demonstrate that miR-27a is an androgen-regulated oncomir in prostate cancer, acting via targeting the tumour suppressor and AR corepressor, Prohibitin (PHB). Increasing miR-27a expression results in reduced PHB mRNA and protein levels, and increased expression of AR target genes and prostate cancer cell growth. This involves a novel mechanism for androgen-mediated miR regulation, whereby AR induces a transient increase in miR-23a27a24-2 transcription, but more significantly accelerates processing of the primiR-23a27a24-2 cluster. Androgens therefore regulate miR-27a expression both transcriptionally (via AR binding to the cluster promoter) and post-transcriptionally (accelerating primiR processing to the mature form). We further show that a miR-27a anti-sense oligonucleotide, by opposing the effects of mir-27a, has therapeutic potential in prostate cancer.


Subject(s)
Androgens/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Oncogene Proteins/genetics , Prostatic Neoplasms/genetics , RNA Processing, Post-Transcriptional , Repressor Proteins/genetics , Base Sequence , Cell Line, Tumor , Humans , Male , MicroRNAs/metabolism , Molecular Sequence Data , Oncogene Proteins/metabolism , Prohibitins , Prostatic Neoplasms/metabolism , Protein Binding , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Repressor Proteins/metabolism
16.
Sci Rep ; 14(1): 11115, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38750183

ABSTRACT

Androgen deprivation therapy (ADT) is the core treatment for advanced prostate cancer (PCa), with a proven survival benefit. ADT lowers circulating testosterone levels throughout the body, but with it comes a variety of reported side effects including fatigue, muscle wastage, weight gain, hot flushes and importantly cognitive impairment, depression, and mood swings. Testosterone has a key role in brain masculinization, but its direct effects are relatively poorly understood, due both to the brain's extreme complexity and the fact that some of testosterone activities are driven via local conversion to oestrogen, especially during embryonic development. The exact roles, function, and location of the androgen receptor (AR) in the adult male brain are still being discovered, and therefore the cognitive side effects of ADT may be unrecognized or under-reported. The age of onset of several neurological diseases overlap with PCa, therefore, there is a need to separate ADT side effects from such co-morbidities. Here we analysed the activity and expression level of the AR in the adult mouse brain, using an ARE-Luc reporter mouse and immunohistochemical staining for AR in all the key brain regions via coronal slices. We further analysed our data by comparing to the Allen Mouse Brain Atlas. AR-driven luciferase activity and distinct nuclear staining for AR were seen in several key brain areas including the thalamus, hypothalamus, olfactory bulb, cerebral cortex, Purkinje cells of the cerebellum and the hindbrain. We describe and discuss the potential role of AR in these areas, to inform and enable extrapolation to potential side effects of ADT in humans.


Subject(s)
Brain , Receptors, Androgen , Receptors, Androgen/metabolism , Animals , Mice , Brain/metabolism , Brain/drug effects , Male
17.
IEEE Sens Lett ; 7(8): 1-4, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37529707

ABSTRACT

Ion-sensitive field-effect transistors (ISFETs) in combination with unmodified complementary metal oxide semiconductors present a point-of-care platform for clinical diagnostics and prognostics. This work illustrates the sensitive and specific detection of two circulating mRNA markers for prostate cancer, the androgen receptor and the TMPRSS2-ERG fusion using a target-specific loop-mediated isothermal amplification method. TMPRSS2-ERG and androgen receptor RNA were detected down to 3x101 and 5x101 copies respectively in under 30 minutes. Administration of these assays onto the ISFET Lab-on-chip device was successful and the specificity of each marker was corroborated with mRNA extracted from prostate cancer cell lines.

18.
Cell Rep ; 42(4): 112396, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37061917

ABSTRACT

Emerging evidence indicates that metabolic dysregulation drives prostate cancer (PCa) progression and metastasis. AMP-activated protein kinase (AMPK) is a master regulator of metabolism, although its role in PCa remains unclear. Here, we show that genetic and pharmacological activation of AMPK provides a protective effect on PCa progression in vivo. We show that AMPK activation induces PGC1α expression, leading to catabolic metabolic reprogramming of PCa cells. This catabolic state is characterized by increased mitochondrial gene expression, increased fatty acid oxidation, decreased lipogenic potential, decreased cell proliferation, and decreased cell invasiveness. Together, these changes inhibit PCa disease progression. Additionally, we identify a gene network involved in cell cycle regulation that is inhibited by AMPK activation. Strikingly, we show a correlation between this gene network and PGC1α gene expression in human PCa. Taken together, our findings support the use of AMPK activators for clinical treatment of PCa to improve patient outcome.


Subject(s)
AMP-Activated Protein Kinases , Prostatic Neoplasms , Male , Humans , AMP-Activated Protein Kinases/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Lipogenesis , Lipid Metabolism , Prostatic Neoplasms/pathology
19.
J Biol Chem ; 286(20): 17796-808, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21454491

ABSTRACT

The Hairy/Enhancer-of-split-related with YRPW-like motif (HEY) family of proteins are transcriptional repressors and downstream effectors of Notch signaling. We previously reported that HEY1 and HEY2 selectively repress androgen receptor (AR) signaling in mammalian cell lines and have shown that in human tissue HEY1 is excluded from the nuclei in prostate cancer but not benign prostatic hyperplasia. We have now characterized a third member of this family, HEYL, which is a more potent repressor of AR activity. HEYL interacted with and repressed AR activation function-1 domain and competitively inhibited SRC1e activation of AR transcriptional activity. Using a cell line inducibly expressing exogenous HEYL, we showed that HEYL represses endogenous AR-regulated genes and reduces androgen-dependent prostate cancer cell growth. Using a trans-repression assay, we identified both trichostatin-sensitive and -insensitive domains within HEYL; however, analysis of endogenous AR target genes suggested that HEYL represses AR activity through histone deacetylase I/II-independent mechanisms. Immunohistochemical analyses of tissue indicated that, in a fashion similar to that previously reported for HEY1, HEYL is excluded from the nuclei in prostate cancer but not adjacent benign tissue. This suggests that nuclear exclusion of HEY proteins may be an important step in the progression of prostate cancer.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Nucleus/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/genetics , Female , Humans , Male , Prostatic Neoplasms/genetics , Protein Structure, Tertiary , Receptors, Androgen/genetics , Repressor Proteins/genetics
20.
Endocr Oncol ; 2(1): R112-R131, 2022 Jan.
Article in English | MEDLINE | ID: mdl-37435460

ABSTRACT

Androgen signalling, through the transcription factor androgen receptor (AR), is vital to all stages of prostate development and most prostate cancer progression. AR signalling controls differentiation, morphogenesis, and function of the prostate. It also drives proliferation and survival in prostate cancer cells as the tumour progresses; given this importance, it is the main therapeutic target for disseminated disease. AR is also essential in the surrounding stroma, for the embryonic development of the prostate and controlling epithelial glandular development. Stromal AR is also important in cancer initiation, regulating paracrine factors that excite cancer cell proliferation, but lower stromal AR expression correlates with shorter time to progression/worse outcomes. The profile of AR target genes is different between benign and cancerous epithelial cells, between castrate-resistant prostate cancer cells and treatment-naïve cancer cells, between metastatic and primary cancer cells, and between epithelial cells and fibroblasts. This is also true of AR DNA-binding profiles. Potentially regulating the cellular specificity of AR binding and action are pioneer factors and coregulators, which control and influence the ability of AR to bind to chromatin and regulate gene expression. The expression of these factors differs between benign and cancerous cells, as well as throughout disease progression. The expression profile is also different between fibroblast and mesenchymal cell types. The functional importance of coregulators and pioneer factors in androgen signalling makes them attractive therapeutic targets, but given the contextual expression of these factors, it is essential to understand their roles in different cancerous and cell-lineage states.

SELECTION OF CITATIONS
SEARCH DETAIL