Abstract
The androgen receptor (AR) is a ligand-activated transcription factor that drives prostate cancer. Since therapies that target the AR are the mainstay treatment for men with metastatic disease, it is essential to understand the molecular mechanisms underlying oncogenic AR signaling in the prostate. miRNAs are small, non-coding regulators of gene expression that play a key role in prostate cancer and are increasingly recognized as targets or modulators of the AR signaling axis. In this review, we examine the regulation of AR signaling by miRNAs and vice versa and discuss how this interplay influences prostate cancer growth, metastasis and resistance to therapy. Finally, we explore the potential clinical applications of miRNAs implicated in the regulation of AR signaling in this prevalent hormone-driven disease.
Introduction
The androgen receptor (AR) signaling axis is critical for normal prostatic development and maintenance and is the major driver of prostate cancer (PCa) growth and disease progression. Androgens such as testosterone and its more active metabolite, dihydrotestosterone (DHT), bind to the AR in the cytoplasm of prostate epithelial cells, which elicits its translocation into the nucleus. Ligand-activated AR binds to specific DNA sequences termed AR response elements (AREs) in regulatory regions of target genes, resulting in transcriptional activation or, less commonly, repression (Wang et al. 2009). In normal prostate epithelial cells, a key function of AR target genes is to maintain luminal differentiation; by contrast, in malignant prostate cells an aberrant AR transcriptional program promotes cell proliferation and survival (Coutinho et al. 2016).
Given the critical role of AR in PCa, androgen deprivation therapy (ADT) is the primary treatment for metastatic disease (Feldman & Feldman 2001). ADT encompasses multiple agents that: (i) decrease circulating androgen levels by inhibiting pituitary signals that stimulate testicular androgen production, i.e. lutenizing hormone-releasing hormone (LHRH) and gonadotropin-releasing hormone (GnRH) agonists; (ii) diminish biosynthesis of androgens, i.e. inhibitors of androgen biosynthetic enzymes; and/or (iii) inhibit AR activity i.e. agents that block androgen binding to the AR, termed AR antagonists. Unfortunately, these treatments inevitably fail, normally after a period of 2–3 years. The resultant disease is termed castration-resistant prostate cancer (CRPC) (Scher et al. 2004). AR remains the key driver of most cases of CRPC. Maintenance of AR activity despite low levels of circulating androgens can be achieved through amplification or mutation of the AR gene, alternative splicing to produce constitutively active AR splice variants, altered expression of AR co-regulators and/or enhanced intra-tumoral androgen production (Coutinho et al. 2016).
miRNAs are a class of small non-coding RNAs, approximately 22 nt in length, that post-transcriptionally silence target mRNAs. An estimated 60% of all protein-coding mRNAs are targeted by miRNAs (Friedman et al. 2009), highlighting the powerful influence these small transcripts can have on the transcriptome and how it is interpreted by the cell. Primary transcripts of miRNA genes (pri-miRNAs), transcribed primarily by RNA polymerase II, can be encoded by sequences within (intragenic) or outside of (intergenic) protein-coding genes (Lee et al. 2004, Rodriguez et al. 2004). Following transcription of a miRNA gene (or host gene), pri-miRNAs are processed into precursor miRNAs (pre-miRNAs) that fold into a stem loop structure and are exported from the nucleus (Kim 2005). Within the cytoplasm, pre-miRNAs are cleaved into a miRNA duplex that is then separated; one strand is the final mature form of approximately 22 bp and the other strand is degraded (Kim 2005). As part of the RNA-induced silencing complex (RISC) that includes Argonaute (Ago) proteins, miRNAs bind to target mRNAs and promote transcript degradation and/or translational inhibition (Fabian & Sonenberg 2012). miRNAs generally bind to the 3′UTR of their target genes through complementarity with a region called the seed sequence, consisting of nucleotides 2–8 on the 5′ to 3′ ends of a miRNA sequence (Lewis et al. 2005), although an increasing number of studies have also identified functional miRNA recognition sites in the coding and 5′UTR regions of genes (Brummer & Hausser 2014, Zhou & Rigoutsos 2014, Ito et al. 2017, Zhang et al. 2018).
Dysregulated miRNA expression is a hallmark of cancer development and metastasis (Calin & Croce 2006, Jackson et al. 2014). PCa-associated miRNAs were initially reported by Porkka and colleagues who identified differential expression of miRNAs between benign and malignant prostatic tissue (Porkka et al. 2007). miRNAs have since been implicated in all aspects of prostate carcinogenesis and progression to metastatic and therapy-resistant disease (Kojima et al. 2017, Luu et al. 2017). Moreover, since some PCa-associated miRNAs exist stably in the circulation of patients, there is considerable interest in their utility as blood-based biomarkers to improve PCa diagnosis, prognosis and treatment management (Fabris et al. 2016, Kanwal et al. 2017, Matin et al. 2018).
In this review we explore the significance of the interplay between the AR signaling axis and miRNAs as it relates to PCa growth and progression. Finally, we speculate on the potential clinical applications of miRNAs implicated in the regulation of AR.
Regulation of AR by microRNAs
Direct targeting of AR by miRNAs
Mechanisms by which miRNAs influence the AR signaling axis are depicted in Fig. 1. One prominent feature of this interplay is direct targeting of the AR 3′UTR by miRNAs. Indeed, a recent study predicted that the AR 3′UTR is likely to be more heavily regulated by miRNAs than all other PCa driver genes and in the top 5% of regulated genes overall (Hamilton et al. 2016). This elegant work exploited photoactivatable ribonucleoside-enhanced cross-linking immunoprecipitation of the Argonaute protein (Ago-PAR-CLIP) to identify 147 miRNA seed sides corresponding to 71 miRNA families in the AR 3′UTR (Table 1). Importantly, four of the miRNAs identified by Hamilton and colleagues – miR-9, miR-34c, miR-185 and miR-488 – had been previously discovered in two high-throughput screens aimed at identifying AR-targeting miRNAs (Östling et al. 2011, Kumar et al. 2016) (Table 1). In addition to these unbiased approaches, many other studies have characterized specific AR-targeting miRNAs in a more directed manner. Interestingly, in addition to classical targeting of the 3′UTR, miRNA regulation of the AR via 5′UTR (miR-31) and the coding region (miR-421, miR-449a, miR-449b, miR-646, miR-371, miR-193a and miR-9) has also been reported (Östling et al. 2011, Lin et al. 2013, Kumar et al. 2016). A list of putative AR-targeting miRNAs and their putative modes of action is provided in Table 1.
Putative AR- and AR variant-targeting miRNAs.
miRNA | Target | Mechanism of regulation | Mimic and/or inhibitor used | Ago-PAR-CLIP binding site/s in 3′UTRa | Readouts | Reference |
---|---|---|---|---|---|---|
miR-9 | AR | 3′UTR and coding region | Mimic | Yes | AR protein and mRNA, AR luciferase reporter (wild-type only) | Östling et al. (2011), Hamilton et al. (2016), Kumar et al. (2016) |
miR-30b-3p | AR | 3′UTR | Mimic and Inhibitor | AR protein and mRNA, AR 3′UTR luciferase reporter (wild-type and mutant) | Kumar et al. (2016) | |
miR-30c-5p | AR | 3′UTR | Mimic | AR protein, AR 3′UTR luciferase reporter (wild-type) | Kumar et al. (2016) | |
miR-30d-5p | AR | 3′UTR | Mimic and Inhibitor | AR protein and mRNA, AR 3′UTR luciferase reporter (wild-type and mutant) | Kumar et al. (2016) | |
miR-31 | AR | 5′UTR and coding region | Mimic | AR protein, AR 3′UTR luciferase reporter (wild-type and mutant) | Lin et al. (2013) | |
miR-34a | AR | 3′UTR | Mimic | Yes | AR protein and mRNA, AR luciferase reporter (wild-type only) | Östling et al. (2011), Hamilton et al. (2016) |
miR-34c | AR | 3′UTR | Mimic | Yes | AR protein and mRNA, AR luciferase reporter (wild-type only) | Östling et al. (2011), Hamilton et al. (2016), Kumar et al. (2016) |
miR-101-3p | AR and AR-V7 | 3′UTR | Mimic | Yes | AR protein, AR 3′UTR luciferase reporter (wild-type only) | Hamilton et al. (2016), Kumar et al. (2016) |
miR-124-3p | AR, AR-V7, AR-V4 | 3′UTR | Mimic | Yes | AR protein, AR 3′UTR luciferase reporter (wild-type and mutated) | Shi et al. (2013, 2015), Hamilton et al. (2016), Kumar et al. (2016) |
miR-135b | AR and AR-V7 | 3′UTR | Mimic | AR protein and mRNA, AR luciferase reporter (wild-type only) | Östling et al. (2011) | |
miR-145 | AR | 3′UTR | Mimic | AR protein and mRNA | Larne et al. (2015), Hamilton et al. (2016) | |
miR-149-3p | AR | 3′UTR | Mimic | AR protein, AR 3′UTR luciferase reporter (wild-type only) | Kumar et al. (2016) | |
miR-181c-5p | AR-V7 | 3′UTR | Mimic | Yes | AR protein and mRNA, AR luciferase reporter (wild-type and mutated) | Wu et al. (2019) |
miR-193a-3p | AR and AR-V7 | Coding region | Mimic | AR protein, AR 3′UTR luciferase reporter (wild-type only) | Kumar et al. (2016) | |
miR-205 | AR | 3′UTR | Mimic | Yes | AR protein, AR luciferase reporter (wild-type) | Hagman et al. (2013), Hamilton et al. (2016), Kumar et al. (2016) |
miR-297 | AR | 3′UTR | Mimic | AR mRNA, AR luciferase reporter (wild-type) | Fang et al. (2016) | |
miR-298 | AR | 3′UTR | Mimic | AR protein and mRNA, AR luciferase reporter (wild-type only) | Östling et al. (2011) | |
miR-299 | AR | 3′UTR | Mimic | Yes | AR protein and mRNA, AR luciferase reporter (wild-type only) | Östling et al. (2011), Hamilton et al. (2016) |
miR-371 | AR and AR-V7 | Coding region | Mimic | AR protein, AR luciferase reporter (wild-type only) | Östling et al. (2011), Kumar et al. (2016) | |
miR-421 | AR | 3′UTR and coding region | Mimic | Yes | AR protein and mRNA, AR luciferase reporter (wild-type only) | Östling et al. (2011), Hamilton et al. (2016) |
miR-425-5p | AR | 3′UTR | Mimic | AR protein, AR 3′UTR luciferase reporter (wild-type) | Kumar et al. (2016) | |
miR-449a | AR | 3′UTR and coding region | Lentiviral miRNA overexpression | AR protein and mRNA | Zheng et al. (2015) | |
miR-449b | AR | 3′UTR and coding region | Mimic | AR protein, AR 3′UTR luciferase reporter (wild-type) | Östling et al. (2011), Kumar et al. (2016) | |
mir-488 | AR and AR-V7 | 3′UTR | Mimic | Yes | AR protein and mRNA, AR luciferase reporter (wild-type and mutated) | Östling et al. (2011), Sikand et al. (2011), Hamilton et al. (2016), Kumar et al. (2016) |
miR-541 | AR | 3′UTR | Mimic | AR protein and mRNA, AR 3′UTR luciferase reporter (wild-type) | Östling et al. (2011), Kumar et al. (2016) | |
miR-634 | AR | 3′UTR | Mimic | AR protein and mRNA, AR luciferase reporter (wild-type only) | Östling et al. (2011), Kumar et al. (2016) | |
miR-635 | AR | 3′UTR | Mimic | AR protein, AR 3′UTR luciferase reporter (wild-type) | Kumar et al. (2016) | |
miR-646 | AR and AR-V7 | Coding region | Mimic | AR protein, AR 3′UTR luciferase reporter (wild-type) | Kumar et al. (2016) | |
miR-650 | AR | 3′UTR | Mimic | AR protein, AR 3′UTR luciferase reporter (wild-type) | Kumar et al. (2016) | |
miR-654 | AR | 3′UTR | Mimic | AR protein and mRNA, AR luciferase reporter (wild-type only) | Östling et al. (2011), Kumar et al. (2016) |
aAgo-PAR-CLIP, photoactivatable ribonucleoside-enhanced cross-linking immunoprecipitation of the Argonaute protein (Hamilton et al. 2016).
We propose that the biological relevance of these AR-targeting miRNAs can be prioritized using a set of discrete parameters. First, miRNAs that have been identified in multiple studies using multiple in vitro models are likely to have greater biological relevance in PCa. Second, it is known that transfection of cells with miRNA mimics can yield non-physiological miRNA activity; therefore, we prioritize studies that have demonstrated AR targeting using both miRNA mimics and inhibitors. Third, given that prostate tumors are ‘addicted’ to AR (Coutinho et al. 2016), one would expect oncogenic selection pressure to downregulate biologically relevant AR-targeting miRNAs. Indeed, a number of AR-targeting miRNAs have been reported to be downregulated in prostate tumors compared to non-malignant prostate tissues. However, it is worth noting that our own analyses of miRNA expression in The Cancer Genome Atlas (TCGA) and Memorial Sloan-Kettering Cancer Center (MSKCC) (Taylor et al. 2010) often contradict the published associations with cancer (Fig. 2A and B). Finally, it would be expected that biologically relevant miRNAs are inversely correlated with AR protein levels in clinical samples. Indeed, our analysis of the TCGA cohort revealed such a correlation for miR-145, miR-205, miR-34a and miR-31 (Fig. 2C). Using these parameters for prioritization and taking into account both published findings and our new analyses, we have generated a list of candidate AR-targeting miRNAs shown in Table 2, which are ranked by predicted relevance in PCa. We propose that downregulation of at least a subset of these miRNAs is a key enabler of enhanced AR expression and activity in this disease.
Assessing the biological relevance of microRNAs reported to target the androgen receptor.
miRNA | No of studies reporting association between miRNA and AR | Reference (score: 1 if 2 or more references) | Inverse correlation between AR protein and miRNA (score: 1 if yes, 0 if N/A or no) | Downregulated in cancer compared to normal tissues (score: 0.5 for yes in each of MSKCC and TCGA datasets, 0 if no) | Inverse correlation between AR and miR in PCa tissues (score: 1 if yes, 0 if unknown) | miRNA status in PCa tissues (score: 1 if downregulated, 0 if unknown) | Reported role of miRNA in PCa (score: 1 if tumor suppressor, 0 unknown) | Rank (sum of score) | References |
---|---|---|---|---|---|---|---|---|---|
miR-205 | 3 | Hagman et al. (2013), Hamilton et al. (2016), Kumar et al. (2016) | Yes | Downregulated in TCGA and MSKCC | Yes, AR transcript and protein | Downregulated in PCa tissues | Tumor suppressor | 6 | Gandellini et al. (2009), Hagman et al. (2013), Kumar et al. (2016) |
miR-145 | 2 | Larne et al. (2015), Hamilton et al. (2016) | Yes | Downregulated in MSKCC | Yes | Downregulated in PCa metastases | Tumor suppressor | 5.5 | Larne et al. (2015) |
miR-34a | 2 | Östling et al. (2011), Hamilton et al. (2016) | Yes | Downregulated in MSKCC, Upregulated in TCGA | Yes, AR protein | Unknown | Tumor suppressor | 4.5 | Liu et al. (2011), Östling et al. (2011), Leite et al. (2015) |
miR-34c | 4 | Östling et al. (2011), Fang et al. (2016), Hamilton et al. (2016), Kumar et al. (2016) | No | N/A | Yes, AR transcript and protein | Downregulated in PCa tissues | Tumor suppressor | 4 | Hagman et al. (2010), Östling et al. (2011) |
miR-101-3p | 2 | Hamilton et al. (2016), Kumar et al (2016) | N/A | Downregulated in MSKCC, Upregulated in TCGA | Yes, AR protein | Unknown | Tumor suppressor | 3.5 | Cao et al. (2010) |
miR-299-3p | 2 | Östling et al. (2011), Hamilton et al. (2016) | No | N/A | Unknown | Unknown | Unknown | 2 | |
miR-421 | 2 | Östling et al. (2011), Hamilton et al. (2016) | No | N/A | Unknown | Downregulated in PCa tissues | Tumor suppressor | 3 | Meng et al. (2016) |
miR-124-3p | 4 | Shi et al. (2013, 2015), Hamilton et al. (2016), Kumar et al. (2016) | N/A | N/A | Unknown | Unknown | Tumor suppressor | 2 | Shi et al. (2015) |
miR-185 | 4 | Östling et al. (2011), Hamilton et al. (2016), Kumar et al. (2016) | No | Upregulated in TCGA | Unknown | Downregulated in PCa tissues | Tumor suppressor | 2 | Qu et al. (2013) |
miR-371 | 3 | Östling et al. (2011) , Leite et al. (2015), Kumar et al. (2016) | N/A | N/A | Unknown | Downregulated in PCa tissues | Unknown | 2 | Leite et al. (2015) |
mir-488 | 4 | Östling et al. (2011), Sikand et al. (2011), Hamilton et al. (2016), Kumar et al. (2016) | N/A | N/A | Unknown | Unknown | Tumor suppressor | 2 | Sikand et al. (2011) |
miR-449a | 2 | Östling et al. (2011), Zheng et al. (2015) | N/A | N/A | Unknown | Unknown | Tumor suppressor | 2 | Noonan et al. (2009), Zheng et al. (2015) |
miR-297 | 2 | Östling et al. (2011) , Fang et al. (2016) | N/A | N/A | Unknown | Unknown | Unknown | 1 | N/A |
miR-449b | 2 | Östling et al. (2011), Kumar et al. (2016) | N/A | N/A | Unknown | Unknown | Unknown | 1 | N/A |
miR-541 | 2 | Östling et al. (2011), Kumar et al. (2016) | No | N/A | Unknown | Unknown | Unknown | 1 | N/A |
miR-634 | 2 | Östling et al. (2011), Kumar et al. (2016) | N/A | N/A | Unknown | Unknown | Unknown | 1 | N/A |
miR-654 | 2 | Östling et al. (2011), Kumar et al. (2016) | No | N/A | Unknown | Unknown | Unknown | 1 | N/A |
miR-9 | 3 | Östling et al. (2011), Hamilton et al. (2016), Kumar et al. (2016) | N/A | N/A | Unknown | Upregulated in high grade tumors | OncomiR | 1 | Seashols-Williams et al. (2016) |
When considering targeting of AR by miRNAs, it is worth noting that the reference AR 3′UTR sequence is annotated as being 6777 nucleotides in length (NM_000044.4), but other isoforms have been reported. For example, LNCaP cells have been reported to express an AR isoform with the canonical ~6.8 kb 3′UTR and another alternatively spliced version that lacks a 3 kb region in the 3′UTR (Faber et al. 1991). Moreover, Ostling and colleagues delineated a 6680 nucleotide AR 3′UTR in VCaP cells, a sequence that was also identified in LNCaP, LAPC-4, 22Rv1 and MDA-PCa-2b cell line models (Östling et al. 2011). In addition to variation in length, five SNPs were identified in the AR 3′UTR in PCa cell lines (Waltering et al. 2006). Interestingly, SNPs have been reported to influence miRNA targeting of multiple genes in PCa (Stegeman et al. 2015a ,b ), although our analyses indicate that none of the AR 3′UTR SNPs occur within conserved miRNA recognition sequences (not shown). Nevertheless, we believe that reported variations in the length and sequence of the AR 3′UTR warrants further consideration in relation to interplay with miRNAs, especially since 3′UTR sequences are often overlooked in genomic and transcriptomic studies.
Direct targeting of AR splice variants by miRNAs
AR splice variants (ARVs) are truncated isoforms of the AR that lack part or all of the C-terminal ligand-binding domain (LBD) (Antonarakis et al. 2016). ARV mRNAs were first identified in PCa cell lines (Dehm et al. 2008, Guo et al. 2009) but have since been detected in patient specimens including primary tumors and metastases, circulating tumor cells and whole blood (Hu et al. 2009, Hornberg et al. 2011, Antonarakis et al. 2014, Liu et al. 2016). Recent RNA-seq data indicate the presence of at least 16 distinct ARV mRNA species in primary PCa (Cancer Genome Atlas Research Network 2015) and 23 in metastatic CRPC (Robinson et al. 2015). A subset of these ARVs are constitutively active (i.e. they can regulate transcription in the absence of androgen) and are therefore resistant to therapies that target the LBD (Chan et al. 2015). Although the relevance of ARVs in driving the growth of CRPC remains to be definitively proven (Luo et al. 2018), it is worth noting that high expression of certain ARVs, such as AR-V7 and AR-V9, correlates with resistance to AR-targeted therapies and worse survival (Qu et al. 2015, Kohli et al. 2017, He et al. 2018).
Given the clinical relevance of ARVs, it is important to understand how they are regulated by miRNAs in PCa. Of note, ARVs possess 3′UTR sequences distinct from the canonical AR transcript (Hu et al. 2011, Shi et al. 2015) (Fig. 3A). Recent studies have begun to decipher miRNA regulation of ARVs. Interestingly, a number of miRNAs regulate both canonical AR and specific ARVs through different target sites. For example, miR-124 targets the AR transcript via a 3′UTR site that is distinct from another functional targeting site in the 3′UTRs of AR-V3, AR-V4 and AR-V7 (Fig. 3B) (Shi et al. 2013, Shi et al. 2015). Although the complete repertoires of ARV-targeting miRNAs remain to be determined, the identification of miRNAs that target both AR and ARVs via distinct sequences is intriguing and may suggest co-evolution of miRNAs and alternative AR splicing. Another mechanism by which miRNAs target both AR and ARV transcripts is via shared sequences in coding regions, as exemplified by translational regulation of AR and AR-V7 by miR-646, miR-371-3p and miR-193a-3p (Kumar et al. 2016).
Importantly, differences in the 3′UTRs of ARVs and AR suggest that many of the identified AR-targeting miRNAs would not influence ARV expression and vice versa. While this remains to be proven for most miRNAs, a recent study provided proof of concept by showing that miR-181c-5p effectively suppresses AR-V7 expression by targeting its 3′UTR but has no effect on the levels of the prototypical AR (Wu et al. 2019). Interestingly, AR-V7 and ARv567es exhibit greater post-transcriptional stability than AR in CRPC bone metastases (Hornberg et al. 2011); it is tempting to speculate that this may be (at least partly) explained by differential miRNA targeting.
Indirect modulation of AR expression and activity by miRNAs
As well as direct regulation of AR, miRNAs can indirectly influence the expression and activity of AR via multiple mechanisms (Fig. 1), including targeting AR co-regulators. Approximately 200 AR co-regulators – broadly classified as co-activators and co-repressors – have been identified, comprising a highly complex and potent system for shaping AR function in PCa (Liu et al. 2017). The co-repressor small heterodimer partner (SHP) is downregulated by miR-141, which is frequently elevated in PCa, thereby indirectly contributing to activation of AR’s transcriptional activity (Xiao et al. 2012). Another AR corepressor, prohibitin (PHB), is targeted by miR-27a, leading to increased expression of AR target genes and PCa cell growth (Fletcher et al. 2012). Interestingly, miR-27a is an androgen-regulated miRNA, creating a feedback loop that represents a novel mechanism by which AR enhances its own activity.
Concomitant with enhanced miRNA-mediated targeting of AR co-repressors in PCa is the frequent loss or downregulation of miRNAs that target AR co-activators. For example, miR-137 targets a suite of AR co-activators and is progressively lost with tumor grade due to DNA methylation of the MIR137 locus (Nilsson et al. 2015). Loss of miR-331-3p, which targets the ERBB2 (HER2) oncogene, another AR co-activator (Craft et al. 1999), also enhances AR signaling in PCa (Epis et al. 2009). The AR co-activators p300/CBP-associated factor and bromodomain-containing 8 isoform 2 are targeted by miR-17-5p and miR-185, respectively, leading to reduced AR transcriptional activity in PCa cells (Xiao et al. 2012, Jiang et al. 2016). Interestingly, miR-185 also directly targets AR, enabling this miRNA to mediate a dual mode of inhibition of the AR signaling axis (Jiang et al. 2016).
Another mechanism by which miRNAs indirectly influence the AR signaling axis is by targeting factors that regulate AR gene expression. For example, miR-let-7c targets the oncogenic transcription factor Myc, thereby indirectly downregulating AR expression (Nadiminty et al. 2012). Similarly, by directly targeting fibronectin type III domain-containing 1, miR-1207-3p reduces fibronectin 1 and subsequently AR expression (Das et al. 2016). Todorova and colleagues reported that miR-204 targeting of the DNA methyltransferase DNMT1 results in decreased methylation of, and hence, increased transcription from, the AR gene promoter (Todorova et al. 2017). The nuclear matrix protein hnRNPH1, which is upregulated in prostate tumors and promotes the expression of AR and AR-V7, AR transactivation and binding to AREs, is a target of miR-212 (Yang et al. 2016); miR-212 is downregulated in prostate tumors and its ectopic expression in PCa cell lines results in decrease in hnRNPH1 and AR expression (Yang et al. 2016). In summary, targeting AR co-regulators and upstream regulators of AR expression allows miRNAs to exert an additional layer of regulation on this key oncogenic signaling axis (Fig. 1).
Regulation of microRNAs by the AR signaling axis
Direct regulation of miRNA gene expression by AR
The AR can directly regulate the PCa ‘miRNAome’ by binding to androgen response elements (AREs) within cis-regulatory regions that regulate miRNA gene expression (Fig. 4). The integration of AR genome-wide DNA-binding profiles (‘cistromes’) with androgen-regulated transcriptomes has greatly facilitated the identification of direct miRNA target genes (e.g.; Takayama et al. 2011, Pasqualini et al. 2015). This strategy has been used successfully irrespective of whether miRNAs are encoded in an intragenic or intergenic manner (Pasqualini et al. 2015). In general, AR induces oncogenic miRNAs, which act as downstream effectors of AR signaling, and represses tumor suppressor miRNAs (Table 3), although exceptions to this rule exist (see paragraph immediately below). Key oncogenic miRNAs that are directly upregulated by AR’s transcriptional activation function include miR-19a, miR-27a, miR-133b and miR-185-5p, all of which can promote PCa cell growth and/or survival (Mo et al. 2013, Yao et al. 2016), whereas tumor suppressor miRNAs directly repressed by AR include miR-221/222 and miR-421 (Meng et al. 2016, Gui et al. 2017).
MicroRNAs directly regulated by the AR signaling axis.
miRNA | Regulation by AR /androgens | miRNA expression in PCa tissues | Proposed function of miRNA | # of studies reporting association between miRNA and AR | References |
---|---|---|---|---|---|
miR-1 | Induced | Downregulated | Tumor suppressor | 1 | Liu et al. (2015) |
miR-19a | Induced | Upregulated (in CRPC) | OncomiR | 1 | Mo et al. (2013), Lu et al. (2015) |
miR-21 | Induced | Upregulated | OncomiR | 4 | Ribas et al. (2009) , Murata et al. (2010), Takayama et al. (2011) , Waltering et al. (2011) |
miR-22 | Induced | Downregulated | Tumor suppressor | 3 | Murata et al. (2010), Waltering et al. (2011), Pasqualini et al. (2015) |
miR-27a | Induced | Unknown | OncomiR (Fletcher et al. 2012)/Tumor suppressor (Wan et al. 2016) | 3 | Murata et al. (2010), Fletcher et al. (2012), Mo et al. (2013) |
miR-29a | Induced | Downregulated | Tumor suppressor | 3 | Ribas et al. (2009), Waltering et al. (2011), Pasqualini et al. (2015) |
miR-32 | Induced | Upregulated | OncomiR | 2 | Jalava et al. (2012), Waltering et al. (2011) |
miR-99a | Induced (Takayama et al. 2011) / Repressed (Sun et al. 2014) | Downregulated | Tumor suppressor | 2 | Sun et al. (2011, 2014), Takayama et al. (2011) |
miR-125b-2 | Induced Murata et al. (2010); Takayama et al. 2011) / Repressed (Sun et al. 2014) | Unknown | OncomiR | 3 | Shi et al. (2007), Murata et al. (2010), Takayama et al. (2011) , Sun et al. (2014) |
miR-133b | Induced | Unknown | Unknown | 1 | Mo et al. (2013) |
miR-135a | Induced | Downregulated | Tumor suppressor | 1 | Kroiss et al. (2015) |
miR-148a | Induced | Upregulated | OncomiR | 4 | Ribas et al. (2009), Murata et al. (2010), Waltering et al. (2011), Jalava et al. (2012) |
miR-182-5p | Induced | Upregulated | OncomiR | 1 | Yao et al. (2016) |
miR-193a-3p | Induced | Unknown | OncomiR | 2 | Waltering et al. (2011), Jia et al. (2017) |
miR-203 | Induced | Upregulated | Tumor suppressor | 1 | Jalava et al. (2012), Siu et al. (2016) |
miR-221/222 | Repressed | Downregulated / Upregulated | OncomiR | 3 | Takayama et al. (2011), Waltering et al. (2011), Jalava et al. (2012) |
miR-421 | Repressed | Unknown | Tumor suppressor | 1 | Meng et al. (2016) |
miR-4496 | Induced | Unknown | Tumor suppressor | 1 | Wang et al. (2018) |
While AR is the major driver of PCa, it also plays an essential role in normal prostate physiology, where it primarily regulates a differentiative rather than proliferative transcriptional program (Coutinho et al. 2016). This concept may explain the observation that AR also promotes the expression of miRNAs that inhibit proliferation (e.g. miR-101, miR-135a and miR-1, which target the oncogenic factors EZH2, ROCK1/2 and SRC respectively; Cao et al. 2010, Kroiss et al. 2015, Liu et al. 2015) and promote epithelial differentiation (e.g. miR-200b, which targets factors that drive epithelial-mesenchymal transition, such as ZEB1; Williams et al. 2013).
Indirect regulation of miRNA expression and/or activity by the AR signaling axis
Indirect regulation of miRNAs by the AR can occur through a variety of mechanisms (Fig. 4). First, AR appears to play an important regulatory role in miRNA biogenesis (Fletcher et al. 2012) by upregulating expression of DICER (Mo et al. 2013) and also, at least for certain miRNA transcripts, modulating the activity of Drosha (Fletcher et al. 2012). The latter mechanism was elegantly elucidated by Fletcher and colleagues who demonstrated a dual mode by which AR regulates miR-27a: AR induced the expression of the primiR-23a27a24-2 cluster and concomitantly accelerated Drosha-mediated processing of this cluster to generate mature miR-27a (Fletcher et al. 2012). The biological significance of AR’s role in regulating the PCa miRNAome was highlighted by the observation that treatment of LNCaP cells with the AR antagonist enzalutamide results in a ~25% reduction in the number of miRNAs associated with gene 3′UTRs (Hamilton et al. 2016).
AR-mediated regulation of the epigenome – more specifically, interplay between the AR and DNA methylation machinery – also modulates miRNA expression. As an example, by negatively regulating DNMT1, AR elicits hypomethylation and activation of the MIR375 promoter (Chu et al. 2014). Similarly, AR appears to regulate the DNA methylation levels of genomic elements that regulate the expression of miR-22 and miR-29a, although the mechanism by which it achieves this is unclear (Pasqualini et al. 2015).
Androgen receptor-mediated rewiring of microRNA/mRNA transcriptional networks
The androgen-regulated transcriptome has been examined in many different PCa models and contexts (Nelson et al. 2002, Velasco et al. 2004, Ngan et al. 2009, Massie et al. 2011, Pomerantz et al. 2015). However, it is often difficult to identify which of these transcripts are directly regulated by AR, largely due to the fact that AR often binds to enhancers that are distal to its target genes (Wang et al. 2007). Carefully assessing the temporality of androgen-mediated gene regulation (Massie et al. 2011), integrating transcriptomic data with AR cistromes (Pomerantz et al. 2015) and/or using more sophisticated transcriptomic techniques such as global run-on sequencing (GRO-seq) (Toropainen et al. 2016), which is designed to identify nascent transcription, have improved the detection of bona fide AR targets in PCa cell lines and tissues. Nevertheless, accurately differentiating direct vs indirect targets of the AR remains challenging.
We propose that indirect regulation of protein-coding genes by androgens/AR is heavily influenced by the same genes being direct targets of AR-regulated miRNAs. Sun and colleagues explored this concept by evaluating the AR-repressed miR-99a/let7c/125b-2 cluster and discovered significant enrichment of AR-induced genes that are putative targets of these miRNAs (Sun et al. 2014). Importantly, this in silico finding was validated for select candidate targets of each miRNA (Sun et al. 2014). We have expanded on this earlier work by intersecting TargetScan-predicted targets (Agarwal et al. 2015) of AR-induced miRNAs with a panel of androgen-repressed genes, and vice versa (Massie et al. 2011) (Table 4). Androgen-repressed genes are significantly over-represented in the putative targets of certain AR-induced miRNAs, whereas androgen-induced genes are over-represented in the targets of certain AR-repressed miRNAs. Our analysis provides strong evidence that protein-coding genes can be regulated by AR signaling via AR-mediated rewiring of miRNA/mRNA transcriptional networks. This phenomenon should be considered when undertaking genomic analysis of AR activity as it may facilitate the elucidation of direct vs indirect transcriptional regulation.
Intersection between AR regulated miRNA (by binding to ARBS) and AR regulated genes.
miRNA | # of predicted miRNA target genesa | Genes in overlap | P valueb |
---|---|---|---|
Overlap between miRNA upregulated by AR and genes downregulated by AR | |||
miR-19 | 1338 | 43 | 0.027 |
miR-21 | 384 | 13 | 0.127 |
miR-22 | 620 | 22 | 0.040 |
miR-27a | 1421 | 56 | 0.0001 |
miR-29a | 1264 | 34 | 0.245 |
miR-32 | 1041 | 36 | 0.015 |
miR-133b | 711 | 18 | 0.421 |
miR-135a | 847 | 26 | 0.108 |
miR-148a | 802 | 26 | 0.067 |
miR-182-5p | 1329 | 44 | 0.012 |
miR-193a-3p | 286 | 12 | 0.041 |
miR-203 | 960 | 25 | 0.342 |
miR-4496 | 5975 | 168 | 0.004 |
Overlap between miRNA downregulated by AR and genes upregulated by AR | |||
miR-221/222 | 504 | 26 | 0.023 |
miR-421 | 450 | 27 | 0.003 |
aPredicted target genes for each miRNA were downloaded from TargetScan (release 7.2; Agarwal et al. 2015). bP values were calculated using hypergeometric probability tests. Genes in universe = 18,393 i.e. total number of genes in Targetscan database. AR-repressed genes (n = 436) and AR-upregulated genes (n = 625) are from Massie et al. (2011).
Regulation of AR and miRNA expression by feedback loops
Feedback loops are a common feature of the interplay between AR and miRNAs. For example, the MIR21 gene is directly activated by the AR; conversely, miR-21 increases expression of AR in PCa cell lines, potentially by targeting PTEN – although the precise mechanism remains unclear (Ribas et al. 2009, Mishra et al. 2014). Since miR-21 potently stimulates prostate tumor growth and metastasis, this feedback loop is likely to play a key role in PCa progression by maintaining expression and activity of 2 oncogenic factors (i.e. AR and miR-21) (Li et al. 2009, Reis et al. 2012, Bonci et al. 2016). Conversely, the AR-repressed miRNAs miR-31 and miR-421 both target the AR directly (Östling et al. 2011, Lin et al. 2013, Meng et al. 2016). In the case of miR-31, this negative feedback loop is shifted in favor of the AR during disease progression, since DNA hypermethylation and subsequent downregulation of miR-31 is a feature of aggressive prostate tumors (Lin et al. 2013, Meng et al. 2016).
Indirect regulatory loops between miRNAs and the AR also exist. For example, miR-190a is directly repressed via an AR-binding site in its promoter, and in turn it suppresses AR expression and activity by targeting the 3′UTR of YB1 (Xu et al. 2015), a transcription factor that acts to enhance AR gene transcription and also as an AR co-activator (Shiota et al. 2011, Xu et al. 2015).
miRNAs as regulators of therapy resistance in prostate cancer
A key feature of CRPC is the acquisition of miRNA activity that promotes cell survival in androgen-depleted conditions. Given the central role of AR in CRPC, it is not surprising that many of these miRNAs act, at least in part, by enhancing AR expression and/or activity. The feedback loop between miR-21 and AR has already been described earlier; in the context of CRPC, it is worth noting that overexpression of miR-21 alone is sufficient to impart androgen-independent growth (Ribas et al. 2009). Unlike miR-21, miR-221/22 is repressed by AR signaling; however, it plays a similar role in promoting CRPC growth: upon castration, it is immediately upregulated and acts to enhance the expression of key cell cycle genes (Gui et al. 2017). Another example of a CRPC-relevant miRNA that influences AR is miR-125b, which was found to be elevated in androgen-independent derivatives of the LNCaP model (Shi et al. 2007). By targeting the AR co-repressor NCoR2, miR-125b enhances AR signaling and thereby promotes androgen-independent growth (Yang et al. 2012).
Concomitant with gain of oncomiRs in CRPC is loss of miRNAs that inhibit survival, proliferation and other oncogenic properties, including miR-146a, which targets the oncogene Rho-activated protein kinase (Lin et al. 2007, 2008); let-7c, which targets the oncogenes Myc and Lin28 (Nadiminty et al. 2012); miR-145-3p, which targets cell cycle-associated genes (Goto et al. 2017) and the AR-targeting miRNAs miR-30c-5p and 30d-5p (Kumar et al. 2016).
While the aforementioned miRNAs appear to play important roles in the progression of PCa to a castration-resistant state, it is noteworthy that studies evaluating expression of miRNAs in CRPC tissues are poorly concordant. Nevertheless, by comparing miRNAs putatively associated with CRPC in three published genome-wide miRNA profiling studies (Jalava et al. 2012, Goto et al. 2015, Goto et al. 2017) and combining this with our own analysis of the Memorial Sloan-Kettering Cancer Center cohort (Taylor et al. 2010), we have identified a set of miRNAs that are reproducibly dysregulated in CRPC (Table 5). miRNAs that were not reproducibly altered in these studies may be false positives or alternatively may simply reflect differences in experimental design; for example, one study compared miRNA expression between benign prostatic hyperplasia and CRPC (Jalava et al. 2012), while the others compare normal prostate tissues or hormone-naïve PCa with CRPC (Taylor et al. 2010, Goto et al. 2015, 2017). Moreover, only the latest of these employed RNA-seq (Goto et al. 2017), while the earlier studies used PCR arrays (Goto et al. 2015) or miRNA microarrays (Taylor et al. 2010, Jalava et al. 2012).
Dysregulated miRNAs in CRPC.
miRNA | Up/Down | # of studies reporting differential expression | Referencesa |
---|---|---|---|
miR-7 | Upregulated | 2 | Taylor et al. (2010), Jalava et al. (2012) |
miR-18a | Upregulated | 2 | Taylor et al. (2010), Jalava et al. (2012) |
miR-23b | Downregulated | 3 | Taylor et al. (2010), Goto et al. (2015), Goto et al. (2017) |
miR-24 | Downregulated | 4 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2015, 2017) |
miR-25 | Upregulated | 2 | Taylor et al. (2010), Jalava et al. (2012) |
miR-27a | Downregulated | 3 | Taylor et al. (2010), Goto et al. (2015, 2017) |
miR-30a | Downregulated | 3 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2015) |
miR-30b-3p | Upregulated | 2 | Taylor et al. (2010), Jalava et al. (2012) |
miR-30e-3p | Downregulated | 3 | Taylor et al. (2010), Goto et al. (2015, 2017) |
miR-95 | Upregulated | 2 | Taylor et al. (2010), Jalava et al. (2012) |
miR-99a | Downregulated | 4 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2015, 2017) |
miR-100 | Downregulated | 3 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2017) |
miR-125a-5p | Downregulated | 3 | Taylor et al. (2010), Goto et al. (2015, 2017) |
miR-125b | Downregulated | 4 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2015, 2017) |
miR-125b-2 | Downregulated | 3 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2017) |
miR-130b | Upregulated | 2 | Taylor et al. (2010), Jalava et al. (2012) |
miR-133a | Downregulated | 4 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2015, 2017) |
miR-143 | Downregulated | 3 | Taylor et al. (2010), Goto et al. (2015, 2017) |
miR-143 | Downregulated | 3 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2017) |
miR-145 | Downregulated | 3 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2017) |
miR-145a | Downregulated | 3 | Taylor et al. (2010), Goto et al. (2015, 2017) |
miR-150 | Downregulated | 3 | Taylor et al. (2010), Goto et al. (2015, 2017) |
miR-152 | Downregulated | 3 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2015) |
miR-181c | Downregulated | 3 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2017) |
miR-182 | Upregulated | 2 | Taylor et al. (2010), Jalava et al. (2012) |
miR-183 | Upregulated | 2 | Taylor et al. (2010), Jalava et al. (2012) |
miR-185 | Upregulated | 2 | Taylor et al. (2010), Jalava et al. (2012) |
miR-196b | Downregulated | 3 | Taylor et al. (2010), Goto et al. (2015, 2017) |
miR-199a-5p | Downregulated | 3 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2017) |
miR-205 | Downregulated | 4 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2015, 2017) |
miR-214 | Downregulated | 3 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2017) |
miR-221 | Downregulated | 4 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2015, 2017) |
miR-222 | Downregulated/Upregulated | 4 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2015, 2017) |
miR-425 | Upregulated | 2 | Taylor et al. (2010), Jalava et al. (2012) |
miR-452 | Downregulated | 3 | Taylor et al. (2010), Jalava et al. (2012), Goto et al. (2017) |
miR-625 | Upregulated | 2 | Taylor et al. (2010), Jalava et al. (2012) |
aThe studies referred to in the above table encompass 32 CRPC samples (18 metastases and 14 localized CRPC), 109 primary prostate tumors, 7 benign prostatic hyperplasia samples and 38 normal prostate tissues. Downregulated miRNA were listed in Table 5 if they were reproducibly altered in >50% of studies i.e. 3 out of 4; upregulated miRNA were listed if they were reproducibly altered in >50% of studies i.e. 2 out of 2 (Goto et al (2015 and 2017) only report miRNAs downregulated in CRPC, and were therefore excluded when identifying upregulated miRNAs). Comparisons were done between benign prostatic hyperplasia vs CRPC (Jalava et al. 2012) or both normal/primary vs CRPC (Taylor et al. 2010, Goto et al. 2015, 2017).
Clinical potential of AR-associated microRNAs in prostate cancer
AR-associated miRNAs as biomarkers
Circulating miRNAs can be detected in many body fluids – most notably serum, plasma and urine – and have shown promise as biomarkers for PCa diagnosis and prognosis and predicting therapy response. Since this topic has been comprehensively encapsulated in recent reviews (Fabris et al. 2016, Kanwal et al. 2017), here we will briefly touch on the relevance of the AR signaling axis to these putative biomarkers. Several PCa-associated circulating miRNAs are known to be regulated by AR, suggesting that – like the classic PCa marker prostate-specific antigen (Catalona et al. 1991) – their release into circulation is likely increased in PCa due to elevated AR activity. One prominent example is miR-375, which has consistently been identified as a potential marker of PCa, with higher levels in serum or plasma being associated with shorter overall survival and metastasis (Mitchell et al. 2008, Brase et al. 2011, Selth et al. 2012, Huang et al. 2015). AR indirectly promotes miR-375 transcription by suppressing DNA methylation of its promoter (Chu et al. 2014), and we have previously shown that miR-375 levels are positively correlated with androgen signaling in multiple tumor datasets (Selth et al. 2017). The release of miR-375 from LNCaP cells into culture medium is stimulated by DHT (Tiryakioglu et al. 2013, Gezer et al. 2015), suggesting that its levels in the blood of patients would be increased in PCa due to elevated AR activity. Other AR-regulated miRNAs that have been proposed as potential serum- or plasma-based biomarkers of PCa include miR-21 (Zhang et al. 2011), miR-125b (Fredsoe et al. 2018), miR-141 (Gonzales et al. 2011), miR-19a (Stuopelyte et al. 2016), miR-27a (Gao et al. 2018) and miR-221/222 (Santos et al. 2014).
Potentially the most useful application of circulating miRNAs would be in predicting response to therapies for CRPC. Given that an important subset of circulating miRNAs are AR regulated, it is conceivable that one or more of these may be useful in predicting response to AR-targeted therapies. Supporting this concept, elevated circulating miR-141 could predict clinical progression in a small cohort of CRPC patients with a sensitivity of 78.9% and specificity of 68.8%, although the therapies they received were mixed (chemotherapy, hormone therapy, or novel agents such as vaccines and kinase inhibitors) (Gonzales et al. 2011). Additionally, high levels of miR-221 in peripheral blood are predictive of early CRPC development (Santos et al. 2014). These examples suggest that circulating miRNAs could be useful additions to the biomarker armamentarium, although robust retrospective and prospective validation studies are required to move this field forward (Tavallaie et al. 2015, Wang et al. 2016).
Exploiting AR-modulating miRNAs as a therapeutic strategy
miRNA-based therapies are being actively pursued for a multitude of diseases, as eloquently described in several recent reviews (Hong et al. 2016, Van der Ree et al. 2016, 2017). In cancer, such therapies are focused on increasing levels of tumor suppressor miRNAs by delivery of miRNA mimics or decreasing levels of oncogenic miRNAs by delivery of antisense oligonucleotides (termed antimiRs or antagomiRs) (D’Angelo et al. 2016). Since a single miRNA can regulate multiple cancer-related pathways, miRNA-based therapies possess considerable promise. However, like other nucleic acid-based strategies, issues related to stability in biofluids, delivery to the tissue of interest and toxicity of mimics and antimiRs have hindered their clinical development (Rupaimoole & Slack 2017). While recent advances in formulation and delivery have led to miRNA-based therapies being tested in a range of clinical studies (Matin et al. 2016), it is worth noting that none are yet used in the treatment of patients.
With these caveats in mind, the application of a miRNA-based therapy in PCa remains a distant goal. Nevertheless, it is clear that opportunities in this area exist, especially given the importance of the interplay between miRNAs and the AR. For example, miR-34a reduces PCa cell viability, at least in part by targeting AR (Östling et al. 2011), but it also downregulates the expression of >30 oncogenes across multiple oncogenic pathways (Beg et al. 2017). Cancer therapies based on master tumor suppressor miRNAs such as miR-34 are attractive because they would be expected to have high efficacy and at the same time reduce opportunities for resistance to develop. In pre-clinical studies, systemic delivery of a miR-34a mimic to mice harboring orthotopic PCa xenografts led to decreased tumor growth and metastasis (Liu et al. 2011). A liposomal formulation of miR-34a, termed MRX34, was evaluated in a Phase 1 clinical trial of patients with primary liver cancer, advanced solid tumors and hematological malignancies. Unfortunately, this trial was terminated due to immune-related serious adverse side effects (Hong et al. 2016, Van Roosbroeck & Calin 2017). Nevertheless, the promising anti-tumor activity of MRX34 observed in a subset of patients (Beg et al. 2017) provides incentive to continue investigating miRNA-based therapies for cancer. Indeed, new strategies to more precisely deliver miRNA-based payloads to solid tumors are already showing promise: exciting findings from a recent phase I clinical trial in mesothelioma demonstrated that miR-16-loaded, EGFR-targeted ‘minicells’ demonstrated an acceptable safety profile and early signs of activity (van Zandwijk et al. 2017).
As an alternative to therapies based on miRNA mimics or antimiRs, other strategies to regulate miRNA expression can be envisioned. Indeed, epigenetic therapies are being intensively investigated for a multitude of diseases, and we propose that an important aspect of their activity relates to miRNA biology. As specific examples associated with the AR signaling axis, re-activation of miR-124 using DNA demethylating agents or miR-320a with the histone deacetylase inhibitor OBP-801 lead to decreased AR expression and activity in PCa (Shi et al. 2013, Sato et al. 2016).
Summary
miRNAs and the AR are involved in an intricate dance mediated by direct interactions or complex indirect mechanisms involving transcription factors, co-regulators and epigenetic machinery (Figs 1 and 4). Despite the complexity of this interplay, one overarching theme is that miRNAs play a major role in enhancing and maintaining AR activity throughout the course of PCa progression; miRNAs that antagonize AR expression and activity are frequently lost, whereas miRNAs that facilitate AR signaling are frequently gained. This phenomenon may be particularly relevant in CRPC, where miRNAs can play key roles in establishing and maintaining resistance to AR-targeted therapies. With this in mind, and given the continued focus on targeting AR in PCa, we propose that exploitation of miRNAs – potentially as biomarkers or therapeutic targets – warrants further consideration as a strategy to manage and/or treat this common disease. Finally, since AR is a key player and being investigated as a target in other solid tumors, including breast, bladder and hepatocellular, the value of understanding its interplay with miRNAs is increased; indeed, recent studies indicate that such interplay may be crucial to the progression of these other cancer types (Xiong et al. 2017, Yang et al. 2018, Xiao et al. 2019).
Declaration of interest
Wayne Tilley is an Advisory Editor of Endocrine-Related Cancer. Wayne Tilley was not involved in the review or editorial process for this paper, on which he is listed as an author. Luke Selth is an Advisory Editor of Endocrine-Related Cancer. Luke Selth was not involved in the review or editorial process for this paper, on which he is listed as an author. The other authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review.
Funding
This work was supported by funding from the National Health and Medical Research Council of Australia (IDs 1083961 and 1121057 to L A S and W D T).
Acknowledgements
The authors wish to thank Dr John Toubia for providing processed miRNA expression data from The Cancer Genome Atlas cohort. The results published here are in part based on data generated by The Cancer Genome Atlas, established by the National Cancer Institute and the National Human Genome Research Institute, and they are grateful to the specimen donors and relevant research groups associated with this project.
References
Agarwal V, Bell GW, Nam JW & Bartel DP 2015 Predicting effective microRNA target sites in mammalian mRNAs. eLife 4 e05005. (https://doi.org/10.7554/eLife.05005)
Antonarakis ES, Lu C, Wang H, Luber B, Nakazawa M, Roeser JC, Chen Y, Mohammad TA, Chen Y, Fedor HL, et al. 2014 AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. New England Journal of Medicine 371 1028–1038. (https://doi.org/10.1056/NEJMoa1315815)
Antonarakis ES, Armstrong AJ, Dehm SM & Luo J 2016 Androgen receptor variant-driven prostate cancer: clinical implications and therapeutic targeting. Prostate Cancer and Prostatic Diseases 19 231–241. (https://doi.org/10.1038/pcan.2016.17)
Beg MS, Brenner AJ, Sachdev J, Borad M, Kang YK, Stoudemire J, Smith S, Bader AG, Kim S & Hong DS 2017 Phase I study of MRX34, A liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Investigational New Drugs 35 180–188. (https://doi.org/10.1007/s10637-016-0407-y)
Bonci D, Coppola V, Patrizii M, Addario A, Cannistraci A, Francescangeli F, Pecci R, Muto G, Collura D, Bedini R, et al. 2016 A microRNA code for prostate cancer metastasis. Oncogene 35 1180–1192. (https://doi.org/10.1038/onc.2015.176)
Brase JC, Johannes M, Schlomm T, Falth M, Haese A, Steuber T, Beissbarth T, Kuner R & Sultmann H 2011 Circulating miRNAs are correlated with tumor progression in prostate cancer. International Journal of Cancer 128 608–616. (https://doi.org/10.1002/ijc.25376)
Brummer A & Hausser J 2014 MicroRNA binding sites in the coding region of mRNAs: extending the repertoire of post-transcriptional gene regulation. BioEssays 36 617–626. (https://doi.org/10.1002/bies.201300104)
Calin GA & Croce CM 2006 MicroRNA signatures in human cancers. Nature Reviews Cancer 6 857–866. (https://doi.org/10.1038/nrc1997)
Cancer Genome Atlas Research Network 2015 The molecular taxonomy of primary prostate cancer. Cell 163 1011–1025. (https://doi.org/10.1016/j.cell.2015.10.025)
Cao P, Deng Z, Wan M, Huang W, Cramer SD, Xu J, Lei M & Sui G 2010 MicroRNA-101 negatively regulates Ezh2 and its expression is modulated by androgen receptor and HIF-1α/HIF-1β. Molecular Cancer 9 108. (https://doi.org/10.1186/1476-4598-9-108)
Catalona WJ, Smith DS, Ratliff TL, Dodds KM, Coplen DE, Yuan JJ, Petros JA & Andriole GL 1991 Measurement of prostate-specific antigen in serum as a screening test for prostate cancer. New England Journal of Medicine 324 1156–1161. (https://doi.org/10.1056/NEJM199104253241702)
Chan SC, Selth LA, Li Y, Nyquist MD, Miao L, Bradner JE, Raj GV, Tilley WD & Dehm SM 2015 Targeting chromatin binding regulation of constitutively active AR variants to overcome prostate cancer resistance to endocrine-based therapies. Nucleic Acids Research 43 5880–5897. (https://doi.org/10.1093/nar/gkv262)
Chu M, Chang Y, Li P, Guo Y, Zhang K & Gao W 2014 Androgen receptor is negatively correlated with the methylation-mediated transcriptional repression of miR-375 in human prostate cancer cells. Oncology Reports 31 34–40. (https://doi.org/10.3892/or.2013.2810)
Coutinho I, Day TK, Tilley WD & Selth LA 2016 Androgen receptor signaling in castration-resistant prostate cancer: a lesson in persistence. Endocrine-Related Cancer 23 T179–T197. (https://doi.org/10.1530/ERC-16-0422)
Craft N, Shostak Y, Carey M & Sawyers CL 1999 A mechanism for hormone-independent prostate cancer through modulation of androgen receptor signaling by the HER-2/neu tyrosine kinase. Nature Medicine 5 280–285. (https://doi.org/10.1038/6495)
D’Angelo B, Benedetti E, Cimini A & Giordano A 2016 MicroRNAs: a puzzling tool in cancer diagnostics and therapy. Anticancer Research 36 5571–5575. (https://doi.org/10.21873/anticanres.11142)
Das DK, Naidoo M, Ilboudo A, Park JY, Ali T, Krampis K, Robinson BD, Osborne JR & Ogunwobi OO 2016 miR-1207-3p regulates the androgen receptor in prostate cancer via FNDC1/fibronectin. Experimental Cell Research 348 190–200. (https://doi.org/10.1016/j.yexcr.2016.09.021)
Dehm SM, Schmidt LJ, Heemers HV, Vessella RL & Tindall DJ 2008 Splicing of a novel androgen receptor exon generates a constitutively active androgen receptor that mediates prostate cancer therapy resistance. Cancer Research 68 5469–5477. (https://doi.org/10.1158/0008-5472.CAN-08-0594)
Epis MR, Giles KM, Barker A, Kendrick TS & Leedman PJ 2009 miR-331-3p regulates ERBB-2 expression and androgen receptor signaling in prostate cancer. Journal of Biological Chemistry 284 24696–24704. (https://doi.org/10.1074/jbc.M109.030098)
Faber PW, van Rooij HC, van der Korput HA, Baarends WM, Brinkmann AO, Grootegoed JA & Trapman J 1991 Characterization of the human androgen receptor transcription unit. Journal of Biological Chemistry 266 10743–10749.
Fabian MR & Sonenberg N 2012 The mechanics of miRNA-mediated gene silencing: a look under the hood of miRISC. Nature Structural and Molecular Biology 19 586–593. (https://doi.org/10.1038/nsmb.2296)
Fabris L, Ceder Y, Chinnaiyan AM, Jenster GW, Sorensen KD, Tomlins S, Visakorpi T & Calin GA 2016 The potential of microRNAs as prostate cancer biomarkers. European Urology 70 312–322. (https://doi.org/10.1016/j.eururo.2015.12.054)
Fang ZY, Xu C, Li YM, Cai XB, Ren SC, Liu HQ, Wang Y, Wang FB, Chen R, Qu M, et al. 2016 A feed-forward regulatory loop between androgen receptor and PlncRNA-1 promotes prostate cancer progression. Cancer Letters 374 62–74. (https://doi.org/10.1016/j.canlet.2016.01.033)
Feldman BJ & Feldman D 2001 The development of androgen-independent prostate cancer. Nature Reviews Cancer 1 34–45. (https://doi.org/10.1038/35094009)
Fletcher CE, Dart DA, Sita-Lumsden A, Cheng H, Rennie PS & Bevan CL 2012 Androgen-regulated processing of the oncomir MiR-27a, which targets prohibitin in prostate cancer. Human Molecular Genetics 21 3112–3127. (https://doi.org/10.1093/hmg/dds139)
Fredsoe J, Rasmussen AKI, Thomsen AR, Mouritzen P, Hoyer S, Borre M, Orntoft TF & Sorensen KD 2018 Diagnostic and prognostic microRNA biomarkers for prostate cancer in cell-free urine. European Urology Focus 4 825–833. (https://doi.org/10.1016/j.euf.2017.02.018)
Friedman RC, Farh KKH, Burge CB & Bartel DP 2009 Most mammalian mRNAs are conserved targets of microRNAs. Genome Research 19 92–105. (https://doi.org/10.1101/gr.082701.108)
Gandellini P, Folini M, Longoni N, Pennati M, Binda M, Colecchia M, Salvioni R, Supino R, Moretti R, Limonta M, et al. 2009 miR-205 Exerts tumor-suppressive functions in human prostate through down-regulation of protein kinase C epsilon. Cancer Research 69 2287–2295. (https://doi.org/10.1158/0008-5472.CAN-08-2894)
Gao W, Hong Z, Huang H, Zhu A, Lin S, Cheng C, Zhang X, Zou G & Shi Z 2018 miR-27a in serum acts as biomarker for prostate cancer detection and promotes cell proliferation by targeting Sprouty2. Oncology Letters 16 5291–5298. (https://doi.org/10.3892/ol.2018.9274)
Gezer U, Tiryakioglu D, Bilgin E, Dalay N & Holdenrieder S 2015 Androgen stimulation of PCA3 and miR-141 and their release from prostate cancer cells. Cell Journal 16 488–493. (https://doi.org/10.22074/cellj.2015.494)
Gonzales JC, Fink LM, Goodman OB Jr, Symanowski JT, Vogelzang NJ & Ward DC 2011 Comparison of circulating microRNA 141 to circulating tumor cells, lactate dehydrogenase, and prostate-specific antigen for determining treatment response in patients with metastatic prostate cancer. Clinical Genitourinary Cancer 9 39–45. (https://doi.org/10.1016/j.clgc.2011.05.008)
Goto Y, Kojima S, Nishikawa R, Kurozumi A, Kato M, Enokida H, Matsushita R, Yamazaki K, Ishida Y, Nakagawa M, et al. 2015 MicroRNA expression signature of castration-resistant prostate cancer: the microRNA-221/222 cluster functions as a tumour suppressor and disease progression marker. British Journal of Cancer 113 1055–1065. (https://doi.org/10.1038/bjc.2015.300)
Goto Y, Kurozumi A, Arai T, Nohata N, Kojima S, Okato A, Kato M, Yamazaki K, Ishida Y, Naya Y, et al. 2017 Impact of novel miR-145-3p regulatory networks on survival in patients with castration-resistant prostate cancer. British Journal of Cancer 117 409–420. (https://doi.org/10.1038/bjc.2017.191)
Gui B, Hsieh CL, Kantoff PW, Kibel AS & Jia L 2017 Androgen receptor-mediated downregulation of microRNA-221 and-222 in castration-resistant prostate cancer. PLoS ONE 12 e0184166. (https://doi.org/10.1371/journal.pone.0184166)
Guo Z, Yang X, Sun F, Jiang R, Linn DE, Chen H, Chen H, Kong X, Melamed J, Tepper CG, et al. 2009 A novel androgen receptor splice variant is up-regulated during prostate cancer progression and promotes androgen depletion–resistant growth. Cancer Research 69 2305–2313. (https://doi.org/10.1158/0008-5472.CAN-08-3795)
Hagman Z, Larne O, Edsjö A, Bjartell A, Ehrnström RA, Ulmert D, Lilja H & Ceder Y 2010 miR-34c is downregulated in prostate cancer and exerts tumor suppressive functions. International Journal of Cancer 127 2768–2776. (https://doi.org/10.1002/ijc.25269)
Hagman Z, Haflidadottir BS, Ceder JA, Larne O, Bjartell A, Lilja H, Edsjo A & Ceder Y 2013 miR-205 negatively regulates the androgen receptor and is associated with adverse outcome of prostate cancer patients. British Journal of Cancer 108 1668–1676. (https://doi.org/10.1038/bjc.2013.131)
Hamilton MP, Rajapakshe KI, Bader DA, Cerne JZ, Smith EA, Coarfa C, Hartig SM & McGuire SE 2016 The landscape of microRNA targeting in prostate cancer defined by AGO-PAR-CLIP. Neoplasia 18 356–370. (https://doi.org/10.1016/j.neo.2016.04.008)
He Y, Lu J, Ye Z, Hao S, Wang L, Kohli M, Tindall DJ, Li B, Zhu R, Wang L, et al. 2018 Androgen receptor splice variants bind to constitutively open chromatin and promote abiraterone-resistant growth of prostate cancer. Nucleic Acids Research 46 1895–1911. (https://doi.org/10.1093/nar/gkx1306)
Hong DS, Kang YK, Brenner AJ, Sachdev JC, Ejadi S, Borad MJ, Kim TY, Lim HY, Park K, Becerra C, et al. 2016 MRX34, a liposomal miR-34 mimic, in patients with advanced solid tumors: final dose-escalation results from a first-in-human phase I trial of microRNA therapy. Journal of Clinical Oncology 34 2508–2508. (https://doi.org/10.1200/JCO.2016.34.15_suppl.2508)
Hornberg E, Ylitalo EB, Crnalic S, Antti H, Stattin P, Widmark A, Bergh A & Wikstrom P 2011 Expression of androgen receptor splice variants in prostate cancer bone metastases is associated with castration-resistance and short survival. PLoS ONE 6 e19059. (https://doi.org/10.1371/journal.pone.0019059)
Hu R, Dunn TA, Wei S, Isharwal S, Veltri RW, Humphreys E, Han M, Partin AW, Vessella RL, Isaacs WB, et al. 2009 Ligand-independent androgen receptor variants derived from splicing of cryptic exons signify hormone-refractory prostate cancer. Cancer Research 69 16–22. (https://doi.org/10.1158/0008-5472.CAN-08-2764)
Hu R, Isaacs WB & Luo J 2011 A snapshot of the expression signature of androgen receptor splicing variants and their distinctive transcriptional activities. Prostate 71 1656–1667. (https://doi.org/10.1002/pros.21382)
Huang X, Yuan T, Liang M, Du M, Xia S, Dittmar R, Wang D, See W, Costello BA, Quevedo F, et al. 2015 Exosomal miR-1290 and miR-375 as prognostic markers in castration-resistant prostate cancer. European Urology 67 33–41. (https://doi.org/10.1016/j.eururo.2014.07.035)
Ito Y, Inoue A, Seers T, Hato Y, Igarashi A, Toyama T, Taganov KD, Boldin MP & Asahara H 2017 Identification of targets of tumor suppressor microRNA-34a using a reporter library system. PNAS 114 3927–3932. (https://doi.org/10.1073/pnas.1620019114)
Jackson BL, Grabowska A & Ratan HL 2014 MicroRNA in prostate cancer: functional importance and potential as circulating biomarkers. BMC Cancer 14 930. (https://doi.org/10.1186/1471-2407-14-930)
Jalava SE, Urbanucci A, Latonen L, Waltering KK, Sahu B, Janne OA, Seppala J, Lahdesmaki H, Tammela TLJ & Visakorpi T 2012 Androgen-regulated miR-32 targets BTG2 and is overexpressed in castration-resistant prostate cancer. Oncogene 31 4460–4471. (https://doi.org/10.1038/onc.2011.624)
Jia L, Gui B, Zheng D, Decker KF, Tinay I, Tan M, Wang X & Kibel AS 2017 Androgen receptor-regulated miRNA-193a-3p targets AJUBA to promote prostate cancer cell migration. Prostate 77 1000–1011. (https://doi.org/10.1002/pros.23356)
Jiang CY, Ruan Y, Wang XH, Zhao W, Jiang Q, Jing YF, Han BM, Xia SJ & Zhao FJ 2016 MiR-185 attenuates androgen receptor function in prostate cancer indirectly by targeting bromodomain containing 8 isoform 2, an androgen receptor co-activator. Molecular and Cellular Endocrinology 427 13–20. (https://doi.org/10.1016/j.mce.2016.02.023)
Kanwal R, Plaga AR, Liu X, Shukla GC & Gupta S 2017 MicroRNAs in prostate cancer: functional role as biomarkers. Cancer Letters 407 9–20. (https://doi.org/10.1016/j.canlet.2017.08.011)
Kim VN 2005 MicroRNA biogenesis: coordinated cropping and dicing. Nature Reviews Molecular Cell Biology 6 376–385. (https://doi.org/10.1038/nrm1644)
Kohli M, Ho Y, Hillman DW, Van Etten JL, Henzler C, Yang R, Sperger JM, Li Y, Tseng E & Hon T 2017 Androgen receptor variant AR-V9 is co-expressed with AR-V7 in prostate cancer metastases and predicts abiraterone resistance. Clinical Cancer Research 23 4704–4715. (https://doi.org/10.1158/1078-0432.CCR-17-0017)
Kojima S, Goto Y & Naya Y 2017 The roles of microRNAs in the progression of castration-resistant prostate cancer. Journal of Human Genetics 62 25–31. (https://doi.org/10.1038/jhg.2016.69)
Kroiss A, Vincent S, Decaussin-Petrucci M, Meugnier E, Viallet J, Ruffion A, Chalmel F, Samarut J & Allioli N 2015 Androgen-regulated microRNA-135a decreases prostate cancer cell migration and invasion through downregulating ROCK1 and ROCK2. Oncogene 34 2846–2855. (https://doi.org/10.1038/onc.2014.222)
Kumar B, Khaleghzadegan S, Mears B, Hatano K, Kudrolli TA, Chowdhury WH, Yeater DB, Ewing CM, Luo J, Isaacs WB, et al. 2016 Identification of miR-30b-3p and miR-30d-5p as direct regulators of androgen receptor signaling in prostate cancer by complementary functional microRNA library screening. Oncotarget 7 72593–72607. (https://doi.org/10.18632/oncotarget.12241)
Larne O, Hagman Z, Lilja H, Bjartell A, Edsjo A & Ceder Y 2015 miR-145 suppress the androgen receptor in prostate cancer cells and correlates to prostate cancer prognosis. Carcinogenesis 36 858–866. (https://doi.org/10.1093/carcin/bgv063)
Lee Y, Kim M, Han J, Yeom KH, Lee S, Baek SH & Kim VN 2004 MicroRNA genes are transcribed by RNA polymerase II. EMBO Journal 23 4051–4060. (https://doi.org/10.1038/sj.emboj.7600385)
Leite KR, Morais DR, Florez MG, Reis ST, Iscaife A, Viana N, Moura CM, Silva IA, Katz BS, Pontes J Jr, et al. 2015 The role of microRNAs 371 and 34a in androgen receptor control influencing prostate cancer behavior. Urologic Oncology 33 267 e215–e222. (https://doi.org/10.1016/j.urolonc.2015.03.002)
Lewis BP, Burge CB & Bartel DP 2005 Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120 15–20. (https://doi.org/10.1016/j.cell.2004.12.035)
Li T, Li D, Sha J, Sun P & Huang Y 2009 MicroRNA-21 directly targets MARCKS and promotes apoptosis resistance and invasion in prostate cancer cells. Biochemical and Biophysical Research Communications 383 280–285. (https://doi.org/10.1016/j.bbrc.2009.03.077)
Lin SL, Chang D & Ying SY 2007 Hyaluronan stimulates transformation of androgen-independent prostate cancer. Carcinogenesis 28 310–320. (https://doi.org/10.1093/carcin/bgl134)
Lin SL, Chiang A, Chang D & Ying SY 2008 Loss of mir-146a function in hormone-refractory prostate cancer. RNA 14 417–424. (https://doi.org/10.1261/rna.874808)
Lin P-C, Chiu Y-L, Banerjee S, Park K, Mosquera JM, Giannopoulou E, Alves P, Tewari AK, Gerstein MB, Beltran H, et al. 2013 Epigenetic repression of miR-31 disrupts androgen receptor homeostasis and contributes to prostate cancer progression. Cancer Research 73 1232–1244. (https://doi.org/10.1158/0008-5472.CAN-12-2968)
Liu C, Kelnar K, Liu B, Chen X, Calhoun-Davis T, Li H, Patrawala L, Yan H, Jeter C, Honorio S, et al. 2011 The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nature Medicine 17 211–215. (https://doi.org/10.1038/nm.2284)
Liu YN, Yin J, Barrett B, Sheppard-Tillman H, Li D, Casey OM, Fang L, Hynes PG, Ameri AH & Kelly K 2015 Loss of androgen-regulated microRNA 1 activates SRC and promotes prostate cancer bone metastasis. Molecular and Cellular Biology 35 1940–1951. (https://doi.org/10.1128/MCB.00008-15)
Liu XC, Ledet E, Li DY, Dotiwala A, Steinberger A, Feibus A, Li JZ, Qi Y, Silberstein J, Lee B, et al. 2016 A whole blood assay for AR-V7 and AR(v567es) in patients with prostate cancer. Journal of Urology 196 1758–1763. (https://doi.org/10.1016/j.juro.2016.06.095)
Liu S, Kumari S, Hu Q, Senapati D, Venkadakrishnan VB, Wang D, DePriest AD, Schlanger SE, Ben-Salem S, Valenzuela MM, et al. 2017 A comprehensive analysis of coregulator recruitment, androgen receptor function and gene expression in prostate cancer. eLife 6 e28482. (https://doi.org/10.7554/eLife.28482)
Lu K, Liu C, Tao T, Zhang X, Zhang L, Sun C, Wang Y, Chen S, Xu B & Chen M 2015 MicroRNA-19a regulates proliferation and apoptosis of castration-resistant prostate cancer cells by targeting BTG1. FEBS Letters 589 1485–1490. (https://doi.org/10.1016/j.febslet.2015.04.037)
Luo J, Attard G, Balk SP, Bevan C, Burnstein K, Cato L, Cherkasov A, De Bono JS, Dong Y, Gao AC, et al. 2018 Role of androgen receptor variants in prostate cancer: report from the 2017 mission androgen receptor variants meeting. European Urology 73 715–723. (https://doi.org/10.1016/j.eururo.2017.11.038)
Luu HN, Lin HY, Sorensen KD, Ogunwobi OO, Kumar N, Chornokur G, Phelan C, Jones D, Kidd L, Batra J, et al. 2017 miRNAs associated with prostate cancer risk and progression. BMC Urology 17 18. (https://doi.org/10.1186/s12894-017-0206-6)
Massie CE, Lynch A, Ramos-Montoya A, Boren J, Stark R, Fazli L, Warren A, Scott H, Madhu B, Sharma N, et al. 2011 The androgen receptor fuels prostate cancer by regulating central metabolism and biosynthesis. EMBO Journal 30 2719–2733. (https://doi.org/10.1038/emboj.2011.158)
Matin F, Jeet V, Clements JA, Yousef GM & Batra J 2016 MicroRNA theranostics in prostate cancer precision medicine. Clinical Chemistry 62 1318–1333. (https://doi.org/10.1373/clinchem.2015.242800)
Matin F, Jeet V, Moya L, Selth LA, Chambers S, Clements JA & Batra J 2018 2018 A plasma biomarker panel of four microRNAs for the diagnosis of prostate cancer. Scientific Reports 8 6653. (https://doi.org/10.1038/s41598-018-24424-w)
Meng D, Yang S, Wan X, Zhang Y, Huang W, Zhao P, Li T, Wang L, Huang Y, Li T, et al. 2016 A transcriptional target of androgen receptor, miR-421 regulates proliferation and metabolism of prostate cancer cells. International Journal of Biochemistry and Cell Biology 73 30–40. (https://doi.org/10.1016/j.biocel.2016.01.018)
Mishra S, Deng JJ, Gowda PS, Rao MK, Lin CL, Chen CL, Huang T & Sun LZ 2014 Androgen receptor and microRNA-21 axis downregulates transforming growth factor beta receptor II (TGFBR2) expression in prostate cancer. Oncogene 33 4097–4106. (https://doi.org/10.1038/onc.2013.374)
Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O’Briant KC, Allen A, et al. 2008 Circulating microRNAs as stable blood-based markers for cancer detection. PNAS 105 10513–10518. (https://doi.org/10.1073/pnas.0804549105)
Mo WJ, Zhang JY, Li X, Meng DL, Gao Y, Yang S, Wan XC, Zhou CH, Guo FH, Huang Y, et al. 2013 Identification of novel AR-targeted microRNAs mediating androgen signalling through critical pathways to regulate cell viability in prostate cancer. PLoS ONE 8 e56592. (https://doi.org/10.1371/journal.pone.0056592)
Murata T, Takayama K, Katayama S, Urano T, Horie-Inoue K, Ikeda K, Takahashi S, Kawazu C, Hasegawa A, Ouchi Y, et al. 2010 miR-148a is an androgen-responsive microRNA that promotes LNCaP prostate cell growth by repressing its target CAND1 expression. Prostate Cancer and Prostatic Diseases 13 356–361. (https://doi.org/10.1038/pcan.2010.32)
Nadiminty N, Tummala R, Lou W, Zhu Y, Zhang J, Chen X, eVere White RW, Kung HJ, Evans CP & Gao AC 2012 MicroRNA let-7c suppresses androgen receptor expression and activity via regulation of Myc expression in prostate cancer cells. Journal of Biological Chemistry 287 1527–1537. (https://doi.org/10.1074/jbc.M111.278705)
Nelson PS, Clegg N, Arnold H, Ferguson C, Bonham M, White J, Hood L & Lin B 2002 The program of androgen-responsive genes in neoplastic prostate epithelium. PNAS 99 11890–11895. (https://doi.org/10.1073/pnas.182376299)
Ngan S, Stronach EA, Photiou A, Waxman J, Ali S & Buluwela L 2009 Microarray coupled to quantitative RT–PCR analysis of androgen-regulated genes in human LNCaP prostate cancer cells. Oncogene 28 2051–2063. (https://doi.org/10.1038/onc.2009.68)
Nilsson EM, Laursen KB, Whitchurch J, McWilliam A, Odum N, Persson JL, Heery DM, Gudas LJ & Mongan NP 2015 MiR137 is an androgen regulated repressor of an extended network of transcriptional coregulators. Oncotarget 6 35710–35725. (https://doi.org/10.18632/oncotarget.5958)
Noonan EJ, Place RF, Pookot D, Basak S, Whitson JM, Hirata H, Giardina C & Dahiya R 2009 miR-449a targets HDAC-1 and induces growth arrest in prostate cancer. Oncogene 28 1714–1724. (https://doi.org/10.1038/onc.2009.19)
Östling P, Leivonen SK, Aakula A, Kohonen P, Mäkelä R, Hagman Z, Edsjö A, Kangaspeska S, Edgren H, Nicorici D, et al. 2011 Systematic analysis of microRNAs targeting the androgen receptor in prostate cancer cells. Cancer Research 71 1956–1967. (https://doi.org/10.1158/0008-5472.CAN-10-2421)
Pasqualini L, Bu H, Puhr M, Narisu N, Rainer J, Schlick B, Schafer G, Angelova M, Trajanoski Z, Borno ST, et al. 2015 miR-22 and miR-29a are members of the androgen receptor cistrome modulating LAMC1 and Mcl-1 in prostate cancer. Molecular Endocrinology 29 1037–1054. (https://doi.org/10.1210/me.2014-1358)
Pomerantz MM, Li FG, Takeda DY, Lenci R, Chonkar A, Chabot M, Cejas P, Vazquez F, Cook J, Shivdasani RA, et al. 2015 The androgen receptor cistrome is extensively reprogrammed in human prostate tumorigenesis. Nature Genetics 47 1346–1351. (https://doi.org/10.1038/ng.3419)
Porkka KP, Pfeiffer MJ, Waltering KK, Vessella RL, Tammela TL & Visakorpi T 2007 MicroRNA expression profiling in prostate cancer. Cancer Research 67 6130–6135. (https://doi.org/10.1158/0008-5472.CAN-07-0533)
Qu F, Cui X, Hong Y, Wang J, Li Y, Chen L, Liu Y, Gao Y, Xu D & Wang Q 2013 MicroRNA-185 suppresses proliferation, invasion, migration, and tumorigenicity of human prostate cancer cells through targeting androgen receptor. Molecular and Cellular Biochemistry 377 121–130. (https://doi.org/10.1007/s11010-013-1576-z)
Qu Y, Dai B, Ye D, Kong Y, Chang K, Jia Z, Yang X, Zhang H, Zhu Y & Shi G 2015 Constitutively active AR-V7 plays an essential role in the development and progression of castration-resistant prostate cancer. Scientific Reports 5 7654. (https://doi.org/10.1038/srep07654)
Reis ST, Pontes J, Antunes AA, Dall’Oglio MF, Dip N, Passerotti CC, Rossini GA, Morais DR, Nesrallah AJ, Piantino C, et al. 2012 miR-21 may acts as an oncomir by targeting RECK, a matrix metalloproteinase regulator, in prostate cancer. BMC Urology 12 14. (https://doi.org/10.1186/1471-2490-12-14)
Ribas J, Ni X, Haffner M, Wentzel EA, Salmasi AH, Chowdhury WH, Kudrolli TA, Yegnasubramanian S, Luo J, Rodriguez R, et al. 2009 miR-21: an androgen receptor–regulated microRNA that promotes hormone-dependent and hormone-independent prostate cancer growth. Cancer Research 69 7165–7169. (https://doi.org/10.1158/0008-5472.CAN-09-1448)
Robinson D, Van Allen EM, Wu YM, Schultz N, Lonigro RJ, Mosquera JM, Montgomery B, Taplin ME, Pritchard CC, Attard G, et al. 2015 Integrative clinical genomics of advanced prostate cancer. Cell 161 1215–1228. (https://doi.org/10.1016/j.cell.2015.05.001)
Rodriguez A, Griffiths-Jones S, Ashurst JL & Bradley A 2004 Identification of mammalian microRNA host genes and transcription units. Genome Research 14 1902–1910. (https://doi.org/10.1101/gr.2722704)
Rupaimoole R & Slack FJ 2017 MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nature Reviews Drug Discovery 16 203–222. (https://doi.org/10.1038/nrd.2016.246)
Santos JI, Teixeira AL, Dias F, Mauricio J, Lobo F, Morais A & Medeiros R 2014 Influence of peripheral whole-blood microRNA-7 and microRNA-221 high expression levels on the acquisition of castration-resistant prostate cancer: evidences from in vitro and in vivo studies. Tumour Biology 35 7105–7113. (https://doi.org/10.1007/s13277-014-1918-9)
Sato S, Katsushima K, Shinjo K, Hatanaka A, Ohka F, Suzuki S, Naiki-Ito A, Soga N, Takahashi S & Kondo Y 2016 Histone deacetylase inhibition in prostate cancer triggers miR-320-Mediated suppression of the androgen receptor. Cancer Research 76 4192–4204. (https://doi.org/10.1158/0008-5472.CAN-15-3339)
Scher HI, Buchanan G, Gerald W, Butler LM & Tilley WD 2004 Targeting the androgen receptor: improving outcomes for castration-resistant prostate cancer. Endocrine-Related Cancer 11 459–476. (https://doi.org/10.1677/erc.1.00525)
Seashols-Williams SJ, Budd W, Clark GC, Wu Q, Daniel R, Dragoescu E & Zehner E 2016 miR-9 acts as an oncomir in prostate cancer through multiple pathways that drive tumour progression and metastasis. PloS One 11 e0159601. (https://doi.org/10.1371/journal.pone.0159601)
Selth LA, Townley S, Gillis JL, Ochnik AM, Murti K, Macfarlane RJ, Chi KN, Marshall VR, Tilley WD & Butler LM 2012 Discovery of circulating microRNAs associated with human prostate cancer using a mouse model of disease. International Journal of Cancer 131 652–661. (https://doi.org/10.1002/ijc.26405)
Selth LA, Das R, Townley SL, Coutinho I, Hanson AR, Centenera MM, Stylianou N, Sweeney K, Soekmadji C, Jovanovic L, et al. 2017 A ZEB1-miR-375-YAP1 pathway regulates epithelial plasticity in prostate cancer. Oncogene 36 24–34. (https://doi.org/10.1038/onc.2016.185)
Shi XB, Xue L, Yang J, Ma AH, Zhao J, Xu M, Tepper CG, Evans CP, Kung HJ & deVere White RW 2007 An androgen-regulated miRNA suppresses Bak1 expression and induces androgen-independent growth of prostate cancer cells. PNAS 104 19983–19988. (https://doi.org/10.1073/pnas.0706641104)
Shi XB, Xue L, Ma AH, Tepper CG, Gandour-Edwards R, Kung HJ & deVere White RW 2013 Tumor suppressive miR-124 targets androgen receptor and inhibits proliferation of prostate cancer cells. Oncogene 32 4130–4138. (https://doi.org/10.1038/onc.2012.425)
Shi XB, Ma AH, Xue L, Li M, Nguyen HG, Yang JC, Tepper CG, Gandour-Edwards R, Evans CP, Kung HJ, et al. 2015 miR-124 and androgen receptor signaling inhibitors repress prostate cancer growth by downregulating androgen receptor splice variants, EZH2, and Src. Cancer Research 75 5309–5317. (https://doi.org/10.1158/0008-5472.CAN-14-0795)
Shiota M, Takeuchi A, Song Y, Yokomizo A, Kashiwagi E, Uchiumi T, Kuroiwa K, Tatsugami K, Fujimoto N, Oda Y, et al. 2011 Y-box binding protein-1 promotes castration-resistant prostate cancer growth via androgen receptor expression. Endocrine-Related Cancer 18 505–517. (https://doi.org/10.1530/ERC-11-0017)
Sikand , Slaibi JE, Singh R, Slane SD & Shukla GC 2011 miR 488* inhibits androgen receptor expression in prostate carcinoma cells. International Journal of Cancer 129 810–819. (https://doi.org/10.1002/ijc.25753)
Siu , Chen WY, Tsai HY, Yeh HL, Yin JJ, Liu SY & Liu YN 2016 Androgen receptor regulates SRC expression through microRNA-203. Oncotarget 7 25726–25741. (https://doi.org/10.18632/oncotarget.8366)
Stegeman S, Amankwah E, Klein K, O’Mara TA, Kim D, Lin HY, Permuth-Wey J, Sellers TA, Srinivasan S, Eeles R, et al. 2015a A large-scale analysis of genetic variants within putative miRNA binding sites in prostate cancer. Cancer Discovery 5 368–379. (https://doi.org/10.1158/2159-8290.CD-14-1057)
Stegeman S, Moya L, Selth LA, Spurdle AB, Clements JA & Batra J 2015b A genetic variant of MDM4 influences regulation by multiple microRNAs in prostate cancer. Endocrine-Related Cancer 22 265–276. (https://doi.org/10.1530/ERC-15-0013)
Stuopelyte K, Daniunaite K, Jankevicius F & Jarmalaite S 2016 Detection of miRNAs in urine of prostate cancer patients. Medicina 52 116–124. (https://doi.org/10.1016/j.medici.2016.02.007)
Sun D, Lee YS, Malhotra A, Kim HK, Matecic M, Evans C, Jensen RV, Moskaluk CA & Dutta A 2011 miR-99 family of microRNAs suppresses the expression of prostate-specific antigen and prostate cancer cell proliferation. Cancer Research 71 1313–1324. (https://doi.org/10.1158/0008-5472.CAN-10-1031)
Sun D, Layer R, Mueller AC, Cichewicz MA, Negishi M, Paschal BM & Dutta A 2014 Regulation of several androgen-induced genes through the repression of the miR-99a/let-7c/miR-125b-2 miRNA cluster in prostate cancer cells. Oncogene 33 1448–1457. (https://doi.org/10.1038/onc.2013.77)
Takayama K, Tsutsumi S, Katayama S, Okayama T, Horie-Inoue K, Ikeda K, Urano T, Kawazu C, Hasegawa A, Ikeo K, et al. 2011 Integration of cap analysis of gene expression and chromatin immunoprecipitation analysis on array reveals genome-wide androgen receptor signaling in prostate cancer cells. Oncogene 30 619–630. (https://doi.org/10.1038/onc.2010.436)
Tavallaie R, De Almeida SRM & Gooding JJ 2015 Toward biosensors for the detection of circulating microRNA as a cancer biomarker: an overview of the challenges and successes. Wiley Interdisciplinary Reviews: Nanomedicine and Nanobiotechnology 7 580–592. (https://doi.org/10.1002/wnan.1324)
Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, Arora VK, Kaushik P, Cerami E, Reva B, et al. 2010 Integrative genomic profiling of human prostate cancer. Cancer Cell 18 11–22. (https://doi.org/10.1016/j.ccr.2010.05.026)
Tiryakioglu D, Bilgin E, Holdenrieder S, Dalay N & Gezer U 2013 miR-141 and miR‑375 induction and release are different from PSA mRNA and PCA3 upon androgen stimulation of LNCaP cells. Biomedical Reports 1 802–806. (https://doi.org/10.3892/br.2013.135)
Todorova K, Metodiev MV, Metodieva G, Mincheff M, Fernandez N & Hayrabedyan S 2017 Micro-RNA-204 participates in TMPRSS2/ERG regulation and androgen receptor reprogramming in prostate cancer. Hormones and Cancer 8 28–48. (https://doi.org/10.1007/s12672-016-0279-9)
Toropainen S, Niskanen EA, Malinen M, Sutinen P, Kaikkonen MU & Palvimo JJ 2016 Global analysis of transcription in castration-resistant prostate cancer cells uncovers active enhancers and direct androgen receptor targets. Scientific Reports 6 33510. (https://doi.org/10.1038/srep33510)
Van der Ree MH, Van Der Meer AJ, Van Nuenen AC, de Bruijne J, Ottosen S, Janssen HL, Kootstra NA & Reesink HW 2016 Miravirsen dosing in chronic hepatitis C patients results in decreased micro RNA-122 levels without affecting other micro RNA s in plasma. Alimentary Pharmacology and Therapeutics 43 102–113. (https://doi.org/10.1111/apt.13432)
van der Ree MH, de Vree JM, Stelma F, Willemse S, van der Valk M, Rietdijk S, Molenkamp R, Schinkel J, van Nuenen AC, Beuers U, et al. 2017 Safety, tolerability, and antiviral effect of RG-101 in patients with chronic hepatitis C: a phase 1B, double-blind, randomised controlled trial. Lancet 389 709–717. (https://doi.org/10.1016/S0140-6736(16)31715-9)
Van Roosbroeck K & Calin GA 2017 Cancer hallmarks and microRNAs: the therapeutic connection. Advances in Cancer Research 135 119–149.. (https://doi.org/10.1016/bs.acr.2017.06.002)
van Zandwijk N, Pavlakis N, Kao SC, Linton A, Boyer MJ, Clarke S, Huynh Y, Chrzanowska A, Fulham MJ, Bailey DL, et al. 2017 Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: a first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncology 18 1386–1396. (https://doi.org/10.1016/S1470-2045(17)30621-6)
Velasco AM, Gillis KA, Li Y, Brown EL, Sadler TM, Achilleos M, Greenberger LM, Frost P, Bai W & Zhang Y 2004 Identification and validation of novel androgen-regulated genes in prostate cancer. Endocrinology 145 3913–3924. (https://doi.org/10.1210/en.2004-0311)
Waltering KK, Wallen MJ, Tammela TL, Vessella RL & Visakorpi T 2006 Mutation screening of the androgen receptor promoter and untranslated regions in prostate cancer. Prostate 66 1585–1591. (https://doi.org/10.1002/pros.20387)
Waltering KK, Porkka KP, Jalava SE, Urbanucci A, Kohonen PJ, Latonen LM, Kallioniemi OP, Jenster G & Visakorpi T 2011 Androgen regulation of micro-RNAs in prostate cancer. Prostate 71 604–614. (https://doi.org/10.1002/pros.21276)
Wan XC, Huang WH, Yang S, Zhang YL, Zhang P, Kong Z, Li T, Wu H, Jing FX & Li Y 2016 Androgen-induced miR-27A acted as a tumor suppressor by targeting MAP2K4 and mediated prostate cancer progression. International Journal of Biochemistry & Cell Biology 79 249–260. (https://doi.org/10.1016/j.biocel.2016.08.043)
Wang Q, Li W, Liu XS, Carroll JS, Janne OA, Keeton EK, Chinnaiyan AM, Pienta KJ & Brown M 2007 A hierarchical network of transcription factors governs androgen receptor-dependent prostate cancer growth. Molecular Cell 27 380–392. (https://doi.org/10.1016/j.molcel.2007.05.041)
Wang Q, Li W, Zhang Y, Yuan X, Xu K, Yu J, Chen Z, Beroukhim R, Wang H, Lupien M, et al. 2009 Androgen receptor regulates a distinct transcription program in androgen-independent prostate cancer. Cell 138 245–256. (https://doi.org/10.1016/j.cell.2009.04.056)
Wang J, Chen JY & Sen S 2016 MicroRNA as biomarkers and diagnostics. Journal of Cellular Physiology 231 25–30. (https://doi.org/10.1002/jcp.25056)
Wang RF, Wang ZF, Cheng Q, Wang GR & Bai ZM 2018 Androgen receptor suppresses prostate cancer cell invasion via altering the miR-4496/beta-catenin signals. Biochemical and Biophysical Research Communications 504 82–88. (https://doi.org/10.1016/j.bbrc.2018.08.134)
Williams LV, Veliceasa D, Vinokour E & Volpert OV 2013 miR-200b inhibits prostate cancer EMT, growth and metastasis. PLoS ONE 8 e83991. (https://doi.org/10.1371/journal.pone.0083991)
Wu G, Sun Y, Xiang Z, Wang K, Liu B, Xiao G, Niu Y, Wu D & Chang C 2019 Preclinical study using circular RNA 17 and micro RNA 181c-5p to suppress the enzalutamide-resistant prostate cancer progression. Cell Death and Disease 10 37. (https://doi.org/10.1038/s41419-018-1048-1)
Xiao J, Gong AY, Eischeid AN, Chen D, Deng C, Young CY & Chen XM 2012 miR-141 modulates androgen receptor transcriptional activity in human prostate cancer cells through targeting the small heterodimer partner protein. Prostate 72 1514–1522. (https://doi.org/10.1002/pros.22501)
Xiao Y, Sun Y, Liu G, Zhao J, Gao Y, Yeh S, Gong L & Chang C 2019 Androgen receptor (AR)/miR-520f-3p/SOX9 signaling is involved in altering hepatocellular carcinoma (HCC) cell sensitivity to the sorafenib therapy under hypoxia via increasing cancer stem cells phenotype. Cancer Letters 444 175–187. (https://doi.org/10.1016/j.canlet.2018.11.004)
Xiong Y, Wang L, Li Y, Chen M, He W & Qi L 2017 The long non-coding RNA XIST interacted with MiR-124 to modulate bladder cancer growth, invasion and migration by targeting androgen receptor (AR). Cellular Physiology and Biochemistry 43 405–418. (https://doi.org/10.1159/000480419)
Xu S, Wang T, Song W, Jiang T, Zhang F, Yin Y, Jiang SW, Wu K, Yu Z, Wang C, et al. 2015 The inhibitory effects of AR/miR-190a/YB-1 negative feedback loop on prostate cancer and underlying mechanism. Scientific Reports 5 13528. (https://doi.org/10.1038/srep13528)