Abstract
Prostate cancer (PCa) is the most common non-skin cancer in men worldwide, resulting in significant mortality and morbidity. Depending on the grade and stage of the cancer, patients may be given radiation therapy, hormonal therapy, or chemotherapy. However, more than half of these patients develop resistance to treatment, leading to disease progression and metastases, often with lethal conse-quences. MicroRNAs (miRNAs) are short, non-coding RNAs, which regulate numerous physiologi-cal as well as pathological processes, including cancer. miRNAs mediate their regulatory effect pre-dominately by binding to the 3′-untranslated region (UTR) of their target mRNAs. In this review, we will describe the mechanisms by which miRNAs mediate resistance to radiation and drug therapy (i.e. hormone therapy and chemotherapy) in PCa, including control of apoptosis, cell growth and prolifera-tion, autophagy, epithelial-to-mesenchymal transition (EMT), invasion and metastasis, and cancer stem cells (CSCs). Furthermore, we will discuss the utility of circulating miRNAs isolated from different body fluids of prostate cancer patients as non-invasive biomarkers of cancer detection, disease pro-gression, and therapy response. Finally, we will shortlist the candidate miRNAs, which may have a role in drug and radioresistance, that could potentially be used as predictive biomarkers of treatment response.
Keywords: MicroRNAs (miRNAs), biomarkers, prostate cancer, therapy response, radiation and drug therapy, apoptosis
1. INTRODUCTION
Prostate cancer (PCa) is the most common non-skin malignancy affecting men, and the fifth leading cause of cancer related mortality in men [1]. Various treatment options are available, depending on the grade and stage of the tumour. The 5-year survival rate is excellent for patients with localised disease, but is considerably lower for advanced disease. Unfortunately, most patients develop resistance to drugs or radiotherapy, and this type of cancer is often aggressive, and has limited response to current treatment modalities [2, 3].
MicroRNAs (miRNAs) are non-coding ribonucleic acids (RNAs), 19-22 nucleotides long, which bind to the 3′- untranslated region (3′UTR) of their target mRNAs, leading either to mRNA destabilization or inhibition of translation [4-6]. miRNAs can also bind to the 5′UTR of target mRNAs, leading to enhanced translation [6]. Interestingly, each miRNA can target several hundred mRNAs, thus, playing a critical role in multiple physiological processes. Hence, their deregulation can lead to widespread detrimental effects. Studies have implicated various miRNAs in cancer initiation, progression and metastasis, by either acting as anti-neoplastic tumour suppressors (downregulated) or tumour promoting oncomiRs (upregulated), in a number of human malignancies, including PCa (reviewed in [7]). miRNAs have also garnered interest as potential biomarkers of cancer detection, progression, and treatment response [8]. Recently, the focus has shifted to circulating miRNAs, which can be isolated from various body fluids of cancer patients, in a non-invasive way. Advantages of circulating miRNAs as biomarkers include 1) their resistance to ribonuclease degradation and physiological conditions including high pH, incubation at room temperature for 24 hours, and frequent freeze-thawing, and 2) relatively easy quantification by PCR-based techniques [9].
In PCa, aberrant expression of miRNAs correlates with resistance to radiotherapy [10], hormone therapy [11], and chemotherapy [12]. This review will discuss the mechanisms by which miRNAs mediate treatment resistance. We have focussed on the miRNAs shortlisted here as they have been directly implicated in resistance to therapy, and their roles validated mostly by tissue specimen and/or in vivo studies (Table 1). We have specifically tried to incorporate novel miRNAs, that have not been reviewed previously. In the second part of this review, we discuss circulating miRNAs, where we describe various body fluids that can be used to isolate and quantify miRNAs as potential biomarkers of drug- and radioresistance in PCa patients, concluding with the list of shortlisted potential candidates.
Table 1.
miRNAs and their direct or functional known targets in prostate cancer. miRNAs can directly bind to the 3'UTR of their mRNA target to suppress its expression or indirectly by other unknown mechanisms.
| miRNA | Role in | Expression Change | Direct or Functional Targets in Prostate Cancer | References |
|---|---|---|---|---|
| miR-212 | CRPC | Downregulated | Lin28B SIRT1 hnRNPH1 |
[19] [22] [23] |
| miR-185 | CRPC | Downregulated | AR BRD8 ISO2 SREBP-1,-2 |
[26] [27] [28] |
| miR-616 | CRPC | Upregulated | TFPI-2 | [29] |
| miR-221/222 cluster | CRPC | Upregulated* | p27(kip1) SIRT1 Caspase-10 HECTD2, RAB1A Ecm29 |
[39] [40] [41] [42] [43] |
| miR-146a | CRPC | Downregulated | ROCK1 Rac1 |
[46, 48] [49, 50] |
| miR-15a-16 cluster | Chemoresistance | Downregulated | CCND1, WNT3A CDK1, CDK2 CMYB, AR TGFβ & Hh pathway genes |
[55] [56] [59] [60] |
| miR-200c | Chemoresistance | Downregulated | E-cadherin, EpCAM, Vimentin, ZEB1 TUBB-3, ZEB1, E-cadherin, Vimentin |
[63-65] [65, 70] |
| miR-128 | Chemoresistance | Downregulated | ZEB1 BMI-1 |
[72] [73] |
| miR-143 | Chemoresistance | Downregulated | ERK5 HK2 KRAS FNDC3B |
[75, 81] [76] [80] [82] |
| miR-31 | Chemoresistance | Downregulated | E2F6 AR, E2F1, E2F2, EXO1, FOXM1, MCM2 |
[87, 91] [88] |
| miR-34a | Chemoresistance | Downregulated | CD44 LEF1 SIRT1, Bcl-2 BCL2 c-Myc BIRC5, TCF7 |
[92] [93] [94, 99] [95] [96] [97] |
| miR-521 | Radioresistance | Downregulated | CSA | [103] |
| miR-95 | Radioresistance | Upregulated | SGPP1 | [106] |
| miR-106b | Radioresistance | Upregulated | Caspase-7 | [111] |
| miR-320 | CRPC, Chemoresistance |
Downregulated | β-catenin LAMP1 AR |
[116] [117] [118] |
| miR-21 | CRPC, Chemoresistance |
Upregulated | PTEN PDCD4 RECK p57Kip2 |
[122] [125, 128] [129, 130] [131] |
| miR-32 | CRPC, Radioresistance |
Upregulated | BTG2 DAB2IP |
[136] [138] |
| miR-205 | CRPC, Chemoresistance, Radioresistance |
Downregulated | BCL2L2 c-SRC BCL2 IL24, IL32 AR MED1 TP53INP1 |
[87] [140] [141] [142] [143] [145] [147, 148] |
* indicates contradictory evidence of role in prostate cancer.
2. Castration-Resistant Prostate Cancer (CRPC)
Prostate cancer cells need androgens for growth and proliferation, so disruption of androgen receptor (AR) signalling is a primary treatment option. Androgen receptor (AR) is a nuclear family transcription factor present in its inactivated form in the cytoplasm. AR dimerises upon ligand binding, and translocates to the nucleus, transcriptionally activating its target genes, leading to increased cell growth and proliferation [13, 14].
At diagnosis of metastatic disease, most PCa patients are androgen-sensitive, and therefore, androgen deprivation therapy (ADT) is the preferred treatment. ADT is also given as part of neo-adjuvant therapy, prior to primary therapy (radical prostatectomy or curative radiation therapy), to shrink the tumour mass, or as adjuvant systemic therapy for high-risk PCa patients, for whom metastatic disease is eventually a reality. Unfortunately, after initially responding to ADT, most patients develop resistance within 18-24 months, progressing to a more aggressive form of PCa, referred to as castration-resistant prostate cancer (CRPC). The progression of PCa from hormone-sensitive to hormone-resistant state is often accompanied by rising levels of serum prostate specific antigen (PSA) [15]. CPRC was previously designated as androgen-independent prostate cancer (AIPC) or hormone-refractory prostate cancer (HRPC) [11]. However, as ADT-resistant prostate cancers are still sensitive to AR pathway signalling, and respond to the nonsteroidal anti-androgenic drug abiraterone acetate [16], and AR antagonists like enzalutamide [17], the more accurate term is CRPC. In this review, we will use the term CRPC for patient studies, and androgen-independent (AI) and androgen-dependent (AD) when referring to PCa cell lines. There is currently no curative therapy available for CRPC patients [18].
2.1. Key miRNAs Implicated in CRPC
2.1.1. miRNA-212
miR-212 expression is downregulated in prostate tissue and serum samples from cancer patients compared with healthy controls [19]. Using an online tool, TargetScan, which predicts the mRNA targets of miRNAs, Lin28B mRNA was identified as a potential target of miR-212 [19]. This prediction was validated in vitro. Lin28B is an RNA binding protein, which plays an oncogenic role [20], and forms a regulatory loop with miR-212 via the c-Myc protein [21], resulting in increased growth in CRPC. miR-212 also regulates Sirtuin 1 (SIRT1) expression by binding to its 3′UTR, leading to inhibition of starvation induced autophagy, angiogenesis, and cellular senescence [22]. Recently, Yang et al., investigated the causes of significantly higher incidence of PCa in African American men compared with Caucasian American men [23]. Decreased expression of miR-212 and aberrant expression of AR and the splicing regulator heterogenous nuclear ribonucleoprotein H1 (hnRNPH1) were associated with an increased incidence of PCa in African American men.
2.1.2. miRNA-185
miRNA-185 has been implicated many cancers including gastric [24], non-small cell lung cancer (NSCLC) [25], and PCa, where it is downregulated in comparison with non-cancerous cells [26]. miR-185 plays an important role in the transition of androgen-dependent PCa cells to androgen-independent cells by binding to the 3′UTR of AR mRNA and decreasing its expression [26]. Further, miR-185 also binds to the 3′UTR of the AR co-activator, bromodomain containing 8 isoform 2 (BRD8 ISO2), reducing its expression [27]. miR-185 along with miR-342 promotes caspase-dependent apoptosis in PCa cells by inhibiting the expression of an important transcription factor needed for lipogenesis, sterol regulatory element-binding protein-1 (SREBP1), and its downstream targets, fatty acid synthase (FASN) and 3-hydroxy 3-methylglutaryl CoA reductase (HMGCR), thereby impeding the tumorigenic potential of the cells [28]. Disruption of lipogenesis and cholesterogenesis halts tumour progression via inhibition of cell proliferation, migration and invasion in vitro, and regression of tumours in vivo (Fig. 1).
Fig. (1).
miRNAs implicated in CRPC, chemo-, and radio-resistance. miRNAs may be upregulated or downregulated in all three types of resistances. A few miRNAs are common between CRPC and radio-resistance, CRPC and chemo-resistance, and in all three.
2.1.3. miRNA-616
miR-616 is overexpressed in PCa tissue compared to normal and benign prostate hyperplasia (BPH) tissue specimens, and also in androgen-independent (AI) PCa cell lines, but not in androgen-dependent (AD) or normal prostate epithelial cell lines [29]. LNCaP (AD) and 22rv1 (AI) cells are commonly used PCa cell lines. When miR-616 overexpressing LNCaP cells were injected into the nude mice, tumour growth remained unaffected even after bilateral orchiectomy, whereas tumour growth rate reduced for the control mice. 22rv1 cells with repressed miR-616 expression had delayed the tumour onset and cancer growth in vivo, suggesting that higher expression of miR-616 promotes castration-independent cell growth. miR-616 mediated the AI growth of
PCa cells by suppressing the expression of tissue factor pathway inhibitor-2 (TFPI-2), which acts as tumour suppressor in various cancers [30-32] including prostate cancer [33].
2.1.4. miRNA-663
There are conflicting results regarding the role of miR-663 in cancer. It appears to promote tumorigenesis and cell proliferation in nasopharyngeal carcinoma [34], but was reported to act as a tumour suppressor in pancreatic cancer [35], and glioblastoma [36]. In PCa, miR-663 expression level increases progressively from BPH to tumour tissue to CRPC tissue specimens, and AI cell lines [37]. This change in miR-663 expression correlated with poor clinical outcome and cancer recurrence in patients. Furthermore, overexpression of miR-663 promoted cell proliferation, invasion, and neuroendocrine differentiation in AI cells [37]. Increased expression of miR-663 may be due to binding of transcription factor Ets2 to its promoter. Ets2 activates the genes required for malignant transformation of PCa cells [38].
2.1.5. miRNA-221/222 Cluster
Overexpression of miR-221/222 has been associated with the prostate cancer cell progression from AD to AI. This may be mediated by miR-221/222 binding to the 3′UTR of p27/kip1, thereby decreasing p27/kip1 expression [39]. miRs-221/222 enhances cell proliferation [39, 40] and migration [40], but suppresses apoptosis [40, 41], and EMT [42] (Fig. 2). However, not all studies confirm the oncogenic role of miRs-221/222 in PCa. Goto et al., demonstrated that miR-221/222 expression was lost in PCa and CRPC tissues compared with the normal prostate epithelium [43]. Furthermore, loss of miR-221/222 expression in vitro was, in part, responsible for decreased invasive and migratory abilities of PCa cells, which appeared to be mediated by a scaffold protein Ecm29.
Fig. (2).
Mechanisms by which miRNAs mediate their castration-resistant, chemoresistant, and radioresistant activities. These mechanisms include apoptosis, cell growth and proliferation, cancer stem cells (CSCs), autophagy, regulation of epithelial-to-mesenchymal transition, and cellular migration and invasion.
2.1.6. miRNA-146a
Tumour suppressor miR-146a inhibits cancer cell growth, migration and invasion [44, 45]. miR-146a expression is downregulated in AI versus AD PCa cells. In androgen-independent PC-3 cells, overexpression of miR-146a resulted in reduced cell proliferation, invasion, and adhesion. This occurred via suppression of the protein Rho-associated, coiled-coil containing protein kinase 1 (ROCK1) [46], which is a promoter of cancer cell invasion and anchorage-independent growth [47]. Another study confirmed this finding, by demonstrating that miR-146a mediates its caspase-3 dependent anti-apoptotic function in AI cells via binding to the 3′UTR of ROCK1 mRNA [48]. miR-146a also directly targets Rac1, a member of Rho family of small guanosine triphosphatases, leading to inhibition of apoptosis and augmentation of cell proliferation in AI cells [49, 50].
3. Chemoresistance
CRPC patients often develop metastases, particularly to the bones. Docetaxel is the preferred chemotherapeutic drug for such patients, although most develop resistance eventually [51, 52]. Currently, not many effective treatment options are available for docetaxel-resistant patients. Docetaxel acts by binding to microtubules leading to their stabilization, mitotic arrest, resulting in apoptosis [53]. Cabazitaxel, a new generation taxane, was approved recently for patients having docetaxel-resistance [54].
3.1. Key miRNAs Implicated in Chemoresistance
3.1.1. miRNAs-15a/miR-16
miRNAs miR-15a and miR-16 are candidate tumour suppressors, frequently downregulated in tissue specimens from PCa patients [55, 56]. Intriguingly, miR-16 expression (along with other miRNAs) is also decreased in normal prostatic tissue of PCa patients compared with tumour-negative healthy men (low-PSA) and tumour-negative controls (high-PSA), suggesting that this loss of expression occurs early during carcinogenesis [57]. The locus encoding miRNAs-15a and -16 is often homozygously deleted in PCa patients [58], and loss of miR-15a and miR-16 was associated with chemotherapy refractory behaviour of the PCa cells in vitro [55]. miR-15a and miR-16 were shown to promote apoptosis, impede cell proliferation, and reduce invasiveness of cancer cells by binding to the 3′UTRs of oncogenes CCND1 (encoding cyclin D1) and WNT3A (member of the wnt family of cysteine-rich, secretory glycoproteins) [55]. AntagomiRs are synthetic oligonucleotides used for knocking down specific miRNAs in vivo. Silencing of miR-15a and miR-16 in the prostates of Balb/c mice using antagomiRs augmented the invasive and proliferative potential of cancer cells. miR-16 overexpression also inhibited prostate tumour growth and bone metastasis in a PCa xenograft model, by decreasing the expression of cell cycle and apoptosis related genes, including Cyclin D3, CDK1, CDK2, Cks1, TAAC1, and TAAC3 [56]. Additionally, miR-15a impaired cell viability and migration by binding to the transcription factor cMYB and AR [59]. Loss of miR-15 and miR-16, along with the overexpression of miR-21 significantly increased the invasiveness of PCa cells, in addition to elevated likelihood of bone marrow metastasis [60]. Interestingly, levels of miR-15-miR-16 are not only decreased in prostate epithelial cells, but also in the surrounding cancer-associated fibroblasts (CAFs) [61].
3.1.2. miRNA-200c
miRNA-200c belongs to the miR-200 family, which includes miR-200a, miR-200b, and miR-429 clustered on chromosome 1, and miR-200c and miR-141 clustered on chromosome 12. miR-200 family members may control maintenance of epithelial characteristics, and their loss contributes to epithelial-to-mesenchymal transition (EMT) [62]. EMT is important in cancer metastasis and is associated with chemotherapy resistance. Commonly used epithelial markers are epithelial cell adhesion molecule (EpCAM) and E-cadherin, and mesenchymal markers are zinc-finger E-box-binding homeobox 1 (ZEB1), ZEB2, and vimentin. Expression of miR-200c is decreased in PCa cells [63, 64], and exogenous expression of miR-200c in DU145 cells resulted in reduced proliferative, migratory and invasive potential of the cancer cells via inhibition of EMT [65]. In PCa patients, neoadjuvant chemotherapy using docetaxel contributed to loss of E-cadherin and miRNAs -200c and -205, whereas, enforced expression of miR-200c and miR-205 in vitro sensitized cells to docetaxel treatment [63]. Interestingly, levels of EpCAM are decreased in docetaxel-resistant cells in vitro, which were restored when either or both miR-200c and miR-205 were made to express in the resistant cells [64]. Intriguingly, expression of miR-200 family members decreases when cells undergo EMT, and expression is restored during mesenchymal-to-epithelial transition (MET) [62]. Promoter hypermethylation, a mechanism by which genes are silenced [66], was shown to suppress miR-200c levels in PCa [67]. By analysing data from The Cancer Genome Atlas Data Portal, Gu et al., suggested that a panel of tissue-derived miRNAs, including miR-200c, miR-182, and miR-221, could be used as a biomarker for PCa detection [68]. Cancer stem cells (CSCs) or tumour initiating cells may play a pivotal role in drug resistance by various mechanisms including enhanced DNA damage responses, ABC transporter expression, aldehyde dehydrogenase activity, and aberrations in key signal transduction pathways [69]. miR-200c along with miR-34a could modulate the hedgehog signalling pathway in CSCs in response to chemotherapeutic drug paclitaxel, resulting in CSC enrichment [70]. Paclitaxel alone could increase the CSC populations, but a combination of paclitaxel and hedgehog pathway inhibitor drug cyclopamine successfully countered this effect and induced apoptosis.
3.1.3. miRNA-128
Normal prostate cells express more miR-128 than invasive PCa cells [71, 72]. Induced expression of miR-128 in DU145 and LNCaP by miR-128 mimics sensitised cancer cells to the chemotherapeutic drug cisplatin [72]. ZEB1 was identified in silico and validated in vitro as miR-128 target. Decreased miR-128 expression resulted in higher ZEB1 expression, and maybe, responsible for increased invasiveness and chemo-refractory behaviour of PCa cells. A second study found that restoration of miR-128 expression correlated with reduced invasive potential of cancer cells in vitro, and suppressed tumour regeneration in vivo [73]. Tumour suppressive functions of miR-128 may also be mediated by binding to the proto-oncogene BMI-1, which is a regulator of prostate stem cells self-renewal and maintenance [74].
3.1.4. miR-143
miR-143 expression is progressively lost as the grade of PCa advances [75, 76]. Lower miR-143 correlated with enhanced cell growth, migratory and invasive ability of PCa cells in vitro and in vivo [77, 78]. KRAS is a member of well-known Ras family of GTP/GDP binding proteins, which promote AI cancer progression [79]. KRAS is suppressed by miR-143 in PCa cells, inactivating the downstream mitogen-activated protein kinase (MAPK) signalling pathway, and increasing docetaxel sensitivity [80]. The anti-proliferative and anti-migratory effects of miR-143 were a result of miR-143 binding to the 3′UTR of extracellular signal-regulated protein kinase 5 (ERK5) mRNA and limiting its expression [75]. The reverse correlation between miR-143 and ERK5 was confirmed by tissue microarray data from 168 PCa patients [81]. Further, miR-143 promoted apoptosis and G1/S cell cycle arrest, and decreased cell viability via binding to the 3′UTR of hexokinase 2 (HK2), an important enzyme for aerobic glycolysis [76]. However, not all studies have substantiated miR-143 as tumour suppressor in PCa. Using spheres of PC-3 cells as a prostate cancer stem cell model, elevated expression of miR-143 was correlated with increased migration and invasion of CSCs [78]. Additionally, downregulated miR-143 suppressed metastasis in athymic nude mice. Functional studies revealed that fibronectin type III domain containing 3B (FNDC3B), an inducer of EMT, is a direct target of miR-143 [82].
3.1.5. miRNA-31
miR-31 is suppressed in various cancers, including nasopharyngeal [83], liver [84], oesophageal [85], and breast [86]. Normal prostate epithelial cells express high miR-31, whereas prostate tumours and PCa cell lines express low levels of miR-31 [87, 88]. Enforced expression of miR-31 and miR-205 rendered the cells sensitive to docetaxel and cisplatin [87]. Transcription factor E2F6, which inhibits hypoxia [89] and UV-induced apoptosis [90], is a direct target of miR-31. Inverse correlation between miR-31 and E2F6 was confirmed by another study [91], which established that a histone deacetylase inhibitor promoted apoptosis by activating miR-31 and consequently, inhibiting E2F6 expression. Intriguingly, AR and miR-31 mutually repressed each other, with AR binding to the promoter of miR-31 and miR-31 binding to the 3′UTR of AR mRNA, leading to the increased aggressiveness of the PCa cells [88]. Additionally, in tumours, miR-31 expression was suppressed by promoter hypermethylation.
3.1.6. miRNA-34a
Studies using cell lines, xenograft mouse models, and clinical specimens have established that miR-34a acts as a tumour suppressor in prostate cancer [92, 93]. miR-34a mediates its anti-tumourigenic actions by inhibiting cell proliferation [94, 95], inducing apoptosis [96, 97], inhibition of EMT, and suppressing migration and invasion in vitro [96], and impeding tumour growth and metastasis in vivo [98]. Restoration of miR-34a in PC3 cells inhibited cell growth and cell cycle progression, and induced apoptosis, indicating sensitization of cells to chemotherapeutic campothecin, along with growth inhibition and cell cycle arrest [99].
4. Radioresistance
Radiation based treatment or radiotherapy (RT) uses high energy rays or proton beams, which severely damage the DNA of cancer cells and induces apoptosis. Radiotherapy is given to patients as part of curative therapy, as an adjuvant therapy following radical prostatectomy [100], or as palliative therapy to relieve the bone pain, particularly in patients whose cancers have metastasised. Although the response rate to RT is about 60% for patients with localised tumours, up to 45% of patients develop recurrent PCa within 5 years [101, 102].
4.1. Key miRNAs Implicated in Radioresistance
4.1.1. miRNA-521
Josson et al., performed miRNA screening on LNCaP and C4-2 cells four hours after radiation treatment, and found that expression levels of miR-521 decreased considerably [103]. Overexpression using miR-521 mimics sensitised the cells to RT, and miR-521 inhibition conferred resistance to RT. Mechanistic studies revealed cockayne syndrome protein A (CSA), a DNA repair protein, as a potential target of miR-521.
4.1.2. miRNA-95
Elevated levels of miR-95 have been linked to increased cell proliferation in colorectal cancer [104], and NSCLC [105], and are also associated with chemo- and radio-resistance in NSCLC [105]. Next generation sequencing on parental radiosensitive and radioresistant PC3 cells generated by fractionated irradiation, identified miR-95 as increased in radioresistant cells [106]. Overexpression of miR-95 caused increased invasiveness, anchorage independent growth, and increased radioresistance of PC3 cells. In athymic nude mice, increased expression of miR-95 in PC3 cells correlated with quicker growth of the tumour. miR-95 targeted the 3'UTR of sphingosine-1-phosphate phosphatase 1 (SGPP1), as determined by reporter assay [106]. SGPP1 suppresses the invasive and migratory capabilities of cancer cells [107]. Interestingly, radioresistant PC3 cells also showed cross-resistance to commonly used chemotherapeutic drugs docetaxel and cisplatin.
4.1.3. miRNA-106b
miRNA-106b is an oncomiR in various cancers including hepatocellular carcinoma [108], cervical carcinoma [109], and colorectal cancer [110]. Recently, miR-106b overexpression in prostate tumours was linked with disease recurrence and metastasis, and shown to directly target caspase-7 [111]. Using a miRNA microarray on samples from LNCaP cells following irradiation, a number of miRNAs were found to be differentially expressed including miR-106b [112]. Increased expression of miR-106b by transient transfection with pre-miR-106b resulted in the suppression of cell cycle inhibitor p21 post radiation, causing G2/M arrest.
5. MiRNAs common in CRPC and Chemoresistance
5.1. miRNA-320
miR-320 acts as a tumour suppressor in cancers, including osteosarcoma [113], glioma [114], and cervical cancer [115]. In PCa, miR-320 is lowly expressed in tumour tissues compared with normal prostate epithelium [116-118], and its overexpression resulted in reduced tumorigenic potential of PCa cells, both in vitro and in vivo [116]. Expression of miR-320 is further downregulated in tissue specimens from CRPC patients. miR-320 directly targeted lysosomal-associated membrane protein 1 (LAMP1) [117], which has been previously associated with tumour invasion and metastasis [119]. Knockdown of miR-320 in PCa cell lines increases the resistance to chemotherapy via enriching the CD44high CSC population [116]. Moreover, miR-320 mediates CSC inhibition by binding to 3′UTR of another target β-catenin. β-catenin is critical for self-renewal and maintenance of stem-like characteristics of the CSCs [120]. In PCa cell lines, histone deacetylase (HDAC) inhibitor OBP-801 reduced AR expression and tumour cell growth, by upregulating miR-320 expression [118]. miR-320 expression also increased when PCa xenograft rats were treated with OBP-801, resulting in decreased tumourigenicity.
5.2. miRNA-21
OncomiR miR-21 has been implicated in numerous solid and haematological malignancies (reviewed in [121]). miR-21 is highly expressed in PCa tissues compared to normal prostate epithelia [122, 123]. Increased expression of miR-21 correlated with cancer recurrence in PCa patients following radical prostatectomy. Upregulated miR-21 correlated with more robust AD and AI growth of PCa cells in vitro and in vivo [124], and conferred resistance to docetaxel in PC-3 cells, conversely, miR-21 knockdown sensitized cells to docetaxel-induced apoptosis [125]. This oncogenic activity of miR-21 was mediated by binding to programmed cell death 4 (PDCD4), a tumour suppressor gene. PDCD4 is a direct target of miR-21 in breast [126], colorectal cancer [127], and PCa [128]. miR-21 promotes cell invasion via suppression of reversion-inducing cysteine-rich protein with Kazul motif (RECK), a matrix metalloproteinase inhibitor [129, 130]. Furthermore, by targeting the coding region of p57Kip2, a cyclin-dependent kinase inhibitor, miR-21 promotes cancer cell migration and anchorage-independent growth in PC-3 and 22rv1 cells [131].
6. MiRNAs common in CRPC and Radioresistance
6.1. miRNA-32
miR-32 was predicted to act as a tumour suppressor in multiple neoplasias, including NSCLC [132, 133], and gastric cancer [134]. In contrast, in colorectal cancer, miR-32 promoted cell growth, migration, and invasion by targeting PTEN [135]. miR-32 expression is regulated by androgen in PCa, and is increased in CRPC tissue specimens compared to PCa and benign hyperplasia samples [136]. LNCaP cells transfected with pre-miR-32 had reduced apoptosis compared to controls, and B-cell translocation gene 2 (BTG2) was identified as a target of miR-32 by mRNA microarray analysis [136]. Loss of BTG2 correlates with the oncogenic transformation of non-tumorigenic PCa cells [137], and a shorter progression free survival time in patients. miR-32 also binds to the 3′UTR to inhibit tumour suppressor DAB2IP [138]. Loss of DAB2IP induced EMT in vitro and promoted distant organ metastases in a xenograft mouse model [139]. Overexpressing miR-32 in PCa cell lines increased radioreistance, whereas its silencing sensitised the cells to radiation treatment. miR-32 appeared to mediate this effect via suppressing DAB2IP mediated autophagy [138].
7. MIRNAS common in CRPC, Chemoresistance, and Radioresistance
7.1. miRNA-205
miR-205 suppresses the growth of tumours in various malignancies, including PCa, where it targets c-SRC to limit growth [140], and Bcl2 and Bcl-w to promote apoptosis [87, 141]. miR-205 expression levels are significantly downregulated in PCa cells compared to normal cells [64, 142-144]. This downregulation can be due to hypermethylation of the miR-205 promoter [87, 145]. Interestingly, miR-205 is a target of tumour suppressor p63, which binds to the miR-205 promoter at two different sites [146]. Enhanced expression of miR-205 results in reduced cell migratory ability via inhibition of EMT. Recently, miR-205 was found play a part in irradiation-induced autophagy, possibly mediated by TP53INP1 [147, 148].
8. Circulating miRNAs as non-invasive biomarkers
The presence of circulating miRNAs in cancer patients was first demonstrated using the sera of Diffuse Large B-cell Lymphoma patients [149]. Building on this, Mitchell et al., demonstrated the utility of circulating miRNAs as blood-based diagnostic tools using plasma and serum samples from PCa patients [9]. Since then, many body fluids have been used for the isolation of miRNAs to try and use differentially expressed miRNAs as diagnostic, prognostic, or predictive biomarkers (Table 2). Recent research has suggested that circulating miRNAs may have a role in cell-to-cell communication as they are commonly found to be packaged in exosomes, microvesicles, and apoptotic bodies, and also associated with argonaute 2 protein [150-152].
Table 2.
Circulating miRNAs used as biomarkers of prostate cancer detection, disease progression, and therapy response, isolated from various body fluids.
| Source | Purpose | miRNA Candidates | References | |
|---|---|---|---|---|
| Plasma | Diagnostic (PCa vs healthy) |
miR-141 | [9] | |
| Plasma | Diagnostic (PCa vs healthy), Prognostic (local vs metastatic) |
miR-21, miR-221, miR-141 |
[153] | |
| Plasma | Predictive (treatment response) |
miR-141 | [154, 155] | |
| Plasma Microvesicles | Diagnostic (PCa vs healthy), Prognostic (local vs metastatic) |
miR-107, miR-574-3p, miR-141, miR-375 |
[155] | |
| Plasma | Diagnostic (PCa vs BPH vs healthy) |
miR-let-7c, miR-let-7e, miR-30c, miR-622, miR-1285 | [156] | |
| Plasma | Prognostic (low vs high-risk) |
miR-20a, miR-21, miR-145, miR-221 | [157] | |
| Plasma | Prognostic (local vs mCRPC) |
miR-16, miR-141, miR-151-3p | [158] | |
| Plasma exosomal RNA | Predictive (drug response) |
miR-1290, miR-375 | [159] | |
| Plasma | Predictive (drug response) |
miR-20a, -20b, -21,-25,-132,-146a,-200a,-200b,-200c,-201b,-222,-375,-429,-590-5p | [160] | |
| Serum | Diagnostic (PCa vs healthy) |
miR-16, miR-34b, miR-92a, miR-92b, miR-103, miR-107, miR-197, miR-328, miR-485-3p, miR-486-5p, miR-574-3p, miR-636, miR-640, miR-766, mi-R885-5p | [161] | |
| Serum | Diagnostic (PCa vs BPH vs healthy) |
let-7i, miR-26a, miR-32, miR-195 | [162] | |
| Serum | Diagnostic (PCa vs healthy) |
miR-24, miR-26b, miR-30c, miR-93, miR-106a, miR-223, miR-451, miR-874, miR-1207-5p, miR-1274a | [163] | |
| Serum | Diagnostic (BPH vs PCa) |
19 differentially expressed miRNAs | [164] | |
| Serum | Prognostic (low vs high-grade) |
miR-141, miR-200b, miR-375 | [165] | |
| Serum | Prognostic (local vs metastatic), Predictive (drug response) |
miR-21 | [166] | |
| Serum | Prognostic (PCa vs mCRPC), Predictive (treatment response) |
miR-141, miR-200a, miR-200c, miR-210, miR-375 miR-210 |
[167] | |
| Serum | Prognostic (low-risk vs mCRPC) |
miR-141, miR-375, miR-378a-5p, miR-409-3p | [168] | |
| Serum | Predictive (biochemical recurrence) |
miR-141, miR-146b-3p, miR-194 | [170] | |
| Serum | Prognostic (BPH or low-grade vs high-grade) |
miR-let-7a, miR-24, miR-26b, miR-30c, miR-93, miR-100, miR-103, miR-106a, miR-107, miR-130b, miR-146a, miR-223, miR-451, miR-874 | [171] | |
| Urine | Diagnostic (PCa vs healthy) |
miR-107, miR-574-3p | [155] | |
| Source | Purpose | miRNA Candidates | References | |
| Urine | Diagnostic (PCa vs healthy) |
miR-205, miR-214 | [172] | |
| Urine | Diagnostic (PCa vs healthy) |
miR-205, miR-183 | [173] | |
| Urine | Diagnostic (PCa vs BPH vs healthy) |
miR-1825, miR-484 | [174] | |
| Urine | Diagnostic (PCa vs healthy) |
miR-187 | [175] | |
| Urine | Diagnostic (PCa vs healthy) |
miR-483-5p | [176] | |
| Seminal Fluid | Diagnostic (PCa vs healthy), Prognostic (low vs high-grade) |
miR-200b, miR-200c, miR-30a, miR-375, miR-99a miR-200b |
[177] | |
| Seminal Fluid | Diagnostic (PCa vs non-PCa) |
miR-200c, miR-125b | [178] | |
Currently, serum PSA levels are used to monitor therapy response in PCa patients. This approach has shortcomings, for example, rising PSA is not always indicative of drug resistance or cancer recurrence. Some patients experience increased PSA levels when starting docetaxel treatment, even if they respond well, whereas, PSA levels may plunge in response to docetaxel in others. Interestingly, PSA levels do not show any increase even after local recurrence. There is a need for novel biomarkers that are better at predicting the treatment response. Here, we will briefly describe the potential biological sources and candidate miRNAs, which were expressed differentially between cancer vs non-cancer patients.
8.1. Plasma
Isolating tumour-derived miRNAs from plasma was first demonstrated using NOD/SCID mice, where expression of miR-629-3p and miR-600, could distinguish between the xenografted mice and controls [9]. Besides, they also measured the expression of six candidate miRNAs (miR-100, miR-125b, miR-141, miR-143, miR-205, and miR-296) in the sera of PCa patients and normal controls, and observed increased miR-141 levels in PCa patients. In another study, increased levels of miR-21 and miR-221 differentiated between localised PCa patients and healthy controls [153]. In the same study, while comparing patients with localised or advanced and metastatic disease, miR-141 emerged as the best indicator of cancer progression. The finding was corroborated by other studies [154, 155], and ability of miR-141 to predict cancer progression was equivalent to other biomarkers including serum PSA, circulating tumour cells (CTCs), and lactate dehydrogenase [154]. An Exiqon miRNA qPCR panel was used to profile 742 miRNAs in microvesicles from the plasma of localised (n=55), and metastatic patients (n=16), and healthy controls (n=28) [155]. Differentially expressed miRNAs included miR-141, miR-375, miR-107, and miR-574-3p. Additionally, microvesicle
and exosome derived miR-375 and miR-141 levels increased in patients with recurrent PCa after surgery compared to patients who did not relapse. To differentiate PCa (n=21) from BPH (n=17) patients, a screen of 754 miRNAs using Illumina Human v2 microarrays was performed [156], and identified candidates (miR-let-7c, miR-let-7e, miR-30c, miR-622, and miR-1285) were validated by qRT-PCR in a large cohort, including PCa (n=80), BPH (n=40), and healthy controls (n=54). Distinct expression patterns of miRNAs miR-20a, miR-21, miR-145, and miR-221 separated high-risk from low-risk patients [157]. miR-141, miR-151-3p, and miR-16 together could accurately distinguish between localised PCa and mCRPC with an improved sensitivity and specificity [158]. More recently, miR-375 and miR-1290 overexpression correlated with poor overall survival in CRPC patients [159]. miRNA profiling of CRPC patients before and after docetaxel chemotherapy yielded a number of differentially expressed miRNAs [160].
8.2. Serum
Besides plasma, serum is used for the isolation of miRNAs for the purpose of diagnosis [161-164] and predicting progression and [165], therapy response [166]. Elevated serum levels of miR-375, miR-141, and miR-200b distinguished between patients with low-grade (Gleason Score 6) and high-grade (Gleason Score 7) cancer, more efficiently than the currently used biomarker, PSA [165]. Increased miR-21 in the sera of CRPC patients, was particularly prominent for patients resistant to docetaxel [166]. To identify prognostic and predictive biomarkers, Cheng et al., used TaqMan Low-Density Array to screen the miRNAs from the sera of mCRPC patients (n=25), and healthy controls (n=25) [167], and Nguyen et al., carried out TaqMan miRNA array using the sera from mCRPC (n=26) and localised PCa patients (n=28) [168]. Both studies found increased expression of miR-375 and miR-141 in the serum samples of mCRPC patients, as well as other distinctly expressed miRNAs, including miR-200a, miR-200c, miR-210, miR-378a-5p, and miR-409-3p. miRNAs have also been used to predict treatment response in PCa, with increased expression of miR-141, miR-146b-3p, and miR-194 predictive of biochemical recurrence, which has been described as the PSA value of at least 0.4 ng/ml followed by another increase [169] in patients post radical prostatectomy [170]. To identify miRNAs that distinguish aggressive from indolent PCa, and predict biochemical recurrence, Mihelich et al, performed qRT-PCR analysis of 21 miRNAs from BPH, low- and high-grade PCa patients, and described a panel of 14 miRNAs that were considerably downregulated in high-grade PCa in comparison with BPH and low-grade PCa [171].
8.3. Urine
The expression of miRNAs in urine was measured in patients with and without PCa following a trans-rectal digital massage [155]. Expression of miRNAs miR-107 and miR-574-3p was increased in urine from PCa patients compared to controls. Another study found that downregulation of miR-205 and miR-214 levels could differentiate between healthy controls and PCa patients with 89% sensitivity and 80% specificity [172]. However, levels of miR-205 alone failed to differentiate PCa patients from the controls in a more recent study [173]. The role of urinary miRNAs in distinguishing PCa patients from benign prostate hyperplasia was investigated; two candidate miRNAs (miR-1825 and miR-484) correlated with the development and progression from benign prostate hyperplasia to PCa [174]. Interestingly, combining urinary miR-187 levels with other predictive factors including serum PSA and urinary prostate cancer antigen 3 (PCA3) was superior for predicting disease progression compared to PSA alone [175]. Recently the expression of three miRNAs (miR-483-5p, miR-1275, and miR-1290) was measured in freely voided urine samples [176]. Increased expression of miR-483-5p was observed in PCa patients but not in controls, suggesting that freely voided urine samples could be used for miRNA detection, bypassing the need for a digital rectal examination or prostate massage. However, miR-483-5p alone was not as robust as serum PSA in detecting PCa.
8.4. Seminal Fluid
Recently, Selth et al., looked into the possibility of using miRNAs isolated from the seminal fluid (SF) of prostate cancer patients for diagnostic and prognostic purposes [177]. A series of candidate miRNAs (miR-200b, miR-200c, miR-30a, miR-375, and miR-99a) were better diagnostic tools than serum PSA. Further analysis demonstrated that combining serum PSA with miR-200b levels was better at distinguishing PCa patients than the PSA alone. Additionally, miR-200b could also successfully separate low-risk patients (Gleason score 6) from high-risk patients (Gleason score > 7). The same group has also demonstrated that combining serum PSA with miR-200c and miR-125b expression, which were derived from the non-sperm cell fraction of the seminal fluid improves the specificity of PCa diagnosis compared to PSA levels alone [178].
Conclusion
In prostate cancer, accumulating evidence suggests that aberrant expression of miRNAs contributes to the development of castration-resistance, chemoresistance, and radio-resistance in patients. Many studies have been performed in vitro, in vivo, or on human tissue samples to reach these conclusions.
Circulating miRNAs are promising candidates as non-invasive biomarkers that have been successfully isolated from plasma, serum, urine, and seminal fluid, with varying functional roles, be it as diagnostic, prognostic, or predictive biomarkers. Recently, strong candidates have emerged for use as diagnostic or prognostic markers, including miR-141, miR-21, miR-221, miR-375, miR-205. Others have been proposed as good predictive biomarkers of therapy response, including miR-141, miR-1290, miR-375, miR-200a, -b, and –c, miR-20a, and –b, miR-21, miR-25, miR-132, miR-146a, miR-201b, miR-222, miR-429, and miR-590-5p (from plasma), and miR-21, miR-210, miR-141, miR-146b-3p, and miR-194 (from serum). We have identified a panel of miRNAs that have also been successfully isolated from various body fluids, which play roles in drug- and radio-resistance, and could potentially be used as biomarkers of treatment response, including miR-21, miR-146a, miR-200c, and miR-222 (Fig. 3). These miRNAs are promising candidates because their roles in drug- and radio-resistance have been validated by various tissue-based studies, and they are known to be isolated from body fluids of cancer patients undergoing therapy.
Fig. (3).
miRNAs having a role in CRPC, chemoresistance, and radioresistance, which have evidence of being isolated from various body fluids of prostate cancer patients. Some miRNAs with good evidence for a role in drug- and radioresistance have also been isolated from the body fluids of cancer patients, and as such are good candidate biomarkers to predict patient responses to treatment.
It is interesting to note that some of the miRNAs overlap between diagnostic, prognostic, and predictive categories, including the well-studied miR-141, miR-375, miR-21, and members of miR-200 family. Also worth mentioning is that we did not find any study which examined the potential of urine or seminal fluid miRNA(s) as biomarkers of treatment response. This would be an interesting topic for future research. A panel of miRNAs could be used in place of PSA, or as an adjunct to PSA, to improve its sensitivity and specificity.
Other key miRNAs that have been described here, for their role in drug- and radioresistance, but have not yet been isolated from the body fluids of cancer patients, should be evaluated for their potential as predictive biomarkers, and compared with the currently available and upcoming candidate biomarkers. Circulating miRNA research is still in its nascent stages, and more conclusive studies, with larger patient cohorts will be required to validate the use of circulating miRNAs as biomarkers of treatment response.
Acknowledgements
We would like to thank Dr Renee Richards, University of Queensland Centre for Clinical Research for reviewing the manuscript and providing insightful comments.
Consent for Publication
Not applicable.
Conflict of Interest
The authors declare no conflict of interests. AR is the recipient of the School of Medicine Postgraduate Research Award, Western Sydney University (WSU). TLR holds a Future Research Leader Fellowship from the Cancer Institute New South Wales (CINSW). This work was jointly funded by WSU and CINSW via the Centre for Oncology Education and Research Translation (CONCERT).
References
- 1.Jemal A., Bray F., Center M.M., Ferlay J., Ward E., Forman D. Global cancer statistics. CA Cancer J. Clin. 2011;61:69–90. doi: 10.3322/caac.20107. [DOI] [PubMed] [Google Scholar]
- 2.Malik R., Khan A.P., Asangani I.A., Cieslik M., Prensner J.R., Wang X., Iyer M.K., Jiang X., Borkin D., Escara-Wilke J., Stender R., Wu Y.M., Niknafs Y.S., Jing X., Qiao Y., Palanisamy N., Kunju L.P., Krishnamurthy P.M., Yocum A.K., Mellacheruvu D., Nesvizhskii A.I., Cao X., Dhanasekaran S.M., Feng F.Y., Grembecka J., Cierpicki T., Chinnaiyan A.M. Targeting the MLL complex in castration-resistant prostate cancer. Nat. Med. 2015;21:344–352. doi: 10.1038/nm.3830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Grossfeld G.D., Olumi A.F., Connolly J.A., Chew K., Gibney J., Bhargava V., Waldman F.M., Carroll P.R. Locally recurrent prostate tumors following either radiation therapy or radical prostatectomy have changes in Ki-67 labeling index, p53 and bcl-2 immunoreactivity. J. Urol. 1998;159:1437–1443. doi: 10.1097/00005392-199805000-00004. [DOI] [PubMed] [Google Scholar]
- 4.Lujambio A., Lowe S.W. The microcosmos of cancer. Nature. 2012;482:347–355. doi: 10.1038/nature10888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.He L., Hannon G.J. MicroRNAs: small RNAs with a big role in gene regulation. Nat. Rev. Genet. 2004;5:522–531. doi: 10.1038/nrg1379. [DOI] [PubMed] [Google Scholar]
- 6.Mirnezami A.H., Pickard K., Zhang L., Primrose J.N., Packham G. MicroRNAs: key players in carcinogenesis and novel therapeutic targets. Eur. J. Surg. Oncol. 2009;35:339–347. doi: 10.1016/j.ejso.2008.06.006. [DOI] [PubMed] [Google Scholar]
- 7.Zhang B., Pan X., Cobb G.P., Anderson T.A. MicroRNAs as oncogenes and tumor suppressors. Dev. Biol. 2007;302:1–12. doi: 10.1016/j.ydbio.2006.08.028. [DOI] [PubMed] [Google Scholar]
- 8.Bertoli G., Cava C., Castiglioni I. MicroRNAs: new biomarkers for diagnosis, prognosis, therapy prediction and therapeutic tools for breast cancer. Theranostics. 2015;5:1122–1143. doi: 10.7150/thno.11543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Mitchell P.S., Parkin R.K., Kroh E.M., Fritz B.R., Wyman S.K., Pogosova-Agadjanyan E.L., Peterson A., Noteboom J., O’Briant K.C., Allen A., Lin D.W., Urban N., Drescher C.W., Knudsen B.S., Stirewalt D.L., Gentleman R., Vessella R.L., Nelson P.S., Martin D.B., Tewari M. Circulating microRNAs as stable blood-based markers for cancer detection. Proc. Natl. Acad. Sci. USA. 2008;105:10513–10518. doi: 10.1073/pnas.0804549105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Korpela E., Vesprini D., Liu S.K. MicroRNA in radiotherapy: miRage or miRador? Br. J. Cancer. 2015;112:777–782. doi: 10.1038/bjc.2015.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.ChunJiao S.; Huan, C.; ChaoYang, X.; GuoMei, R. Uncovering the roles of miRNAs and their relationship with androgen receptor in prostate cancer. IUBMB Life. 2014;66:379–386. doi: 10.1002/iub.1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Sun X., Li Y., Yu J., Pei H., Luo P., Zhang J. miR-128 modulates chemosensitivity and invasion of prostate cancer cells through targeting ZEB1. Jpn. J. Clin. Oncol. 2015;45:474–482. doi: 10.1093/jjco/hyv027. [DOI] [PubMed] [Google Scholar]
- 13.Gnanapragasam V.J., Robson C.N., Neal D.E., Leung H.Y. Regulation of FGF8 expression by the androgen receptor in human prostate cancer. Oncogene. 2002;21:5069–5080. doi: 10.1038/sj.onc.1205663. [DOI] [PubMed] [Google Scholar]
- 14.Takayama K., Kaneshiro K., Tsutsumi S., Horie-Inoue K., Ikeda K., Urano T., Ijichi N., Ouchi Y., Shirahige K., Aburatani H., Inoue S. Identification of novel androgen response genes in prostate cancer cells by coupling chromatin immunoprecipitation and genomic microarray analysis. Oncogene. 2007;26:4453–4463. doi: 10.1038/sj.onc.1210229. [DOI] [PubMed] [Google Scholar]
- 15.Scher H.I. Halabi, S.; Tannock, I.; Morris, M.; Sternberg, C.N.; Carducci, M.A.; Eisenberger, M.A.; Higano, C.; Bubley, G.J.; Dreicer, R.; Petrylak, D.; Kantoff, P.; Basch, E.; Kelly, W.K.; Figg, W.D.; Small, E.J.; Beer, T.M.; Wilding, G.; Martin, A.; Hussain, M.; and Prostate Cancer Clinical Trials Working, G. Design and end points of clinical trials for patients with progressive prostate cancer and castrate levels of testosterone: recommendations of the Prostate Cancer Clinical Trials Working Group. J. Clin. Oncol. 2008;26:1148–1159. doi: 10.1200/JCO.2007.12.4487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.de Bono J.S., Logothetis C.J., Molina A., Fizazi K., North S., Chu L., Chi K.N., Jones R.J., Goodman O.B., Jr, Saad F., Staffurth J.N., Mainwaring P., Harland S., Flaig T.W., Hutson T.E., Cheng T., Patterson H., Hainsworth J.D., Ryan C.J., Sternberg C.N., Ellard S.L., Flechon A., Saleh M., Scholz M., Efstathiou E., Zivi A., Bianchini D., Loriot Y., Chieffo N., Kheoh T., Haqq C.M., Scher H.I. Investigators, C.-A.-. Abiraterone and increased survival in metastatic prostate cancer. N. Engl. J. Med. 2011;364:1995–2005. doi: 10.1056/NEJMoa1014618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Beer T.M., Armstrong A.J., Rathkopf D.E., Loriot Y., Sternberg C.N., Higano C.S., Iversen P., Bhattacharya S., Carles J., Chowdhury S., Davis I.D., de Bono J.S., Evans C.P., Fizazi K., Joshua A.M., Kim C.S., Kimura G., Mainwaring P., Mansbach H., Miller K., Noonberg S.B., Perabo F., Phung D., Saad F., Scher H.I., Taplin M.E., Venner P.M., Tombal B., Investigators P. Enzalutamide in metastatic prostate cancer before chemotherapy. N. Engl. J. Med. 2014;371:424–433. doi: 10.1056/NEJMoa1405095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Massard C., Fizazi K. Targeting continued androgen receptor signaling in prostate cancer. Clin. Cancer Res. 2011;17:3876–3883. doi: 10.1158/1078-0432.CCR-10-2815. [DOI] [PubMed] [Google Scholar]
- 19.Borrego-Diaz E., Powers B.C., Azizov V., Lovell S., Reyes R., Chapman B., Tawfik O., McGregor D., Diaz F.J., Wang X., Veldhuizen P.V. A potential regulatory loop between Lin28B: miR212 in androgen-independent prostate cancer. Int. J. Oncol. 2014;45:2421–2429. doi: 10.3892/ijo.2014.2647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Tummala R., Nadiminty N., Lou W., Zhu Y., Gandour-Edwards R., Chen H.W., Evans C.P., Gao A.C. Lin28 promotes growth of prostate cancer cells and activates the androgen receptor. Am. J. Pathol. 2013;183:288–295. doi: 10.1016/j.ajpath.2013.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bernard D., Pourtier-Manzanedo A., Gil J., Beach D.H. Myc confers androgen-independent prostate cancer cell growth. J. Clin. Invest. 2003;112:1724–1731. doi: 10.1172/JCI19035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ramalinga M., Roy A., Srivastava A., Bhattarai A., Harish V., Suy S., Collins S., Kumar D. MicroRNA-212 negatively regulates starvation induced autophagy in prostate cancer cells by inhibiting SIRT1 and is a modulator of angiogenesis and cellular senescence. Oncotarget. 2015;6:34446–34457. doi: 10.18632/oncotarget.5920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Yang Y., Jia D., Kim H., Abd Elmageed Z.Y., Datta A., Davis R., Srivastav S., Moroz K., Crawford B.E., Moparty K., Thomas R., Hudson R.S., Ambs S., Abdel-Mageed A.B. Dysregulation of miR-212 promotes castration resistance through hnRNPH1-mediated regulation of AR and AR-V7: implications for racial disparity of prostate cancer. Clin. Cancer Res. 2016;22:1744–1756. doi: 10.1158/1078-0432.CCR-15-1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Li Q., Wang J.X., He Y.Q., Feng C., Zhang X.J., Sheng J.Q., Li P.F. MicroRNA-185 regulates chemotherapeutic sensitivity in gastric cancer by targeting apoptosis repressor with caspase recruitment domain. Cell Death Dis. 2014;5:e1197. doi: 10.1038/cddis.2014.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Li S., Ma Y., Hou X., Liu Y., Li K., Xu S., Wang J. MiR-185 acts as a tumor suppressor by targeting AKT1 in non-small cell lung cancer cells. Int. J. Clin. Exp. Pathol. 2015;8:11854–11862. [PMC free article] [PubMed] [Google Scholar]
- 26.Qu F., Cui X., Hong Y., Wang J., Li Y., Chen L., Liu Y., Gao Y., Xu D., Wang Q. MicroRNA-185 suppresses proliferation, invasion, migration, and tumorigenicity of human prostate cancer cells through targeting androgen receptor. Mol. Cell. Biochem. 2013;377:121–130. doi: 10.1007/s11010-013-1576-z. [DOI] [PubMed] [Google Scholar]
- 27.Jiang C.Y., Ruan Y., Wang X.H., Zhao W., Jiang Q., Jing Y.F., Han B.M., Xia S.J., Zhao F.J. MiR-185 attenuates androgen receptor function in prostate cancer indirectly by targeting bromodomain containing 8 isoform 2, an androgen receptor co-activator. Mol. Cell. Endocrinol. 2016;427:13–20. doi: 10.1016/j.mce.2016.02.023. [DOI] [PubMed] [Google Scholar]
- 28.Li X., Chen Y.T., Josson S., Mukhopadhyay N.K., Kim J., Freeman M.R., Huang W.C. MicroRNA-185 and 342 inhibit tumorigenicity and induce apoptosis through blockade of the SREBP metabolic pathway in prostate cancer cells. PLoS One. 2013;8:e70987. doi: 10.1371/journal.pone.0070987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ma S., Chan Y.P., Kwan P.S., Lee T.K., Yan M., Tang K.H., Ling M.T., Vielkind J.R., Guan X.Y., Chan K.W. MicroRNA-616 induces androgen-independent growth of prostate cancer cells by suppressing expression of tissue factor pathway inhibitor TFPI-2. Cancer Res. 2011;71:583–592. doi: 10.1158/0008-5472.CAN-10-2587. [DOI] [PubMed] [Google Scholar]
- 30.Sato N., Parker A.R., Fukushima N., Miyagi Y., Iacobuzio-Donahue C.A., Eshleman J.R., Goggins M. Epigenetic inactivation of TFPI-2 as a common mechanism associated with growth and invasion of pancreatic ductal adenocarcinoma. Oncogene. 2005;24:850–858. doi: 10.1038/sj.onc.1208050. [DOI] [PubMed] [Google Scholar]
- 31.Wong C.M., Ng Y.L., Lee J.M., Wong C.C., Cheung O.F., Chan C.Y., Tung E.K., Ching Y.P., Ng I.O. Tissue factor pathway inhibitor-2 as a frequently silenced tumor suppressor gene in hepatocellular carcinoma. Hepatology. 2007;45:1129–1138. doi: 10.1002/hep.21578. [DOI] [PubMed] [Google Scholar]
- 32.Ran Y., Pan J., Hu H., Zhou Z., Sun L., Peng L., Yu L., Sun L., Liu J., Yang Z. A novel role for tissue factor pathway inhibitor-2 in the therapy of human esophageal carcinoma. Hum. Gene Ther. 2009;20:41–49. doi: 10.1089/hum.2008.129. [DOI] [PubMed] [Google Scholar]
- 33.Konduri S.D., Tasiou A., Chandrasekar N., Rao J.S. Overexpression of tissue factor pathway inhibitor-2 (TFPI-2), decreases the invasiveness of prostate cancer cells in vitro. Int. J. Oncol. 2001;18:127–131. [PubMed] [Google Scholar]
- 34.Yi C., Wang Q., Wang L., Huang Y., Li L., Liu L., Zhou X., Xie G., Kang T., Wang H., Zeng M., Ma J., Zeng Y., Yun J.P. MiR-663, a microRNA targeting p21(WAF1/CIP1), promotes the proliferation and tumorigenesis of nasopharyngeal carcinoma. Oncogene. 2012;31:4421–4433. doi: 10.1038/onc.2011.629. [DOI] [PubMed] [Google Scholar]
- 35.Zang W., Wang Y., Wang T., Du Y., Chen X., Li M., Zhao G. miR-663 attenuates tumor growth and invasiveness by targeting eEF1A2 in pancreatic cancer. Mol. Cancer. 2015;14:37. doi: 10.1186/s12943-015-0315-3. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 36.Shi Y., Chen C., Yu S.Z., Liu Q., Rao J., Zhang H.R., Xiao H.L., Fu T.W., Long H., He Z.C., Zhou K., Yao X.H., Cui Y.H., Zhang X., Ping Y.F., Bian X.W. miR-663 Suppresses Oncogenic Function of CXCR4 in Glioblastoma. Clin. Cancer Res. 2015;21:4004–4013. doi: 10.1158/1078-0432.CCR-14-2807. [DOI] [PubMed] [Google Scholar]
- 37.Jiao L., Deng Z., Xu C., Yu Y., Li Y., Yang C., Chen J., Liu Z., Huang G., Li L.C., Sun Y. miR-663 induces castration-resistant prostate cancer transformation and predicts clinical recurrence. J. Cell. Physiol. 2014;229:834–844. doi: 10.1002/jcp.24510. [DOI] [PubMed] [Google Scholar]
- 38.Sementchenko V.I., Schweinfest C.W., Papas T.S., Watson D.K. ETS2 function is required to maintain the transformed state of human prostate cancer cells. Oncogene. 1998;17:2883–2888. doi: 10.1038/sj.onc.1202220. [DOI] [PubMed] [Google Scholar]
- 39.Galardi S., Mercatelli N., Giorda E., Massalini S., Frajese G.V., Ciafre S.A., Farace M.G. miR-221 and miR-222 expression affects the proliferation potential of human prostate carcinoma cell lines by targeting p27Kip1. J. Biol. Chem. 2007;282:23716–23724. doi: 10.1074/jbc.M701805200. [DOI] [PubMed] [Google Scholar]
- 40.Yang X., Yang Y., Gan R., Zhao L., Li W., Zhou H., Wang X., Lu J., Meng Q.H. Down-regulation of mir-221 and mir-222 restrain prostate cancer cell proliferation and migration that is partly mediated by activation of SIRT1. PLoS One. 2014;9:e98833. doi: 10.1371/journal.pone.0098833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Wang L., Liu C., Li C., Xue J., Zhao S., Zhan P., Lin Y., Zhang P., Jiang A., Chen W. Effects of microRNA-221/222 on cell proliferation and apoptosis in prostate cancer cells. Gene. 2015;572:252–258. doi: 10.1016/j.gene.2015.07.017. [DOI] [PubMed] [Google Scholar]
- 42.Sun T., Wang X., He H.H., Sweeney C.J., Liu S.X., Brown M., Balk S., Lee G.S., Kantoff P.W. MiR-221 promotes the development of androgen independence in prostate cancer cells via downregulation of HECTD2 and RAB1A. Oncogene. 2014;33:2790–2800. doi: 10.1038/onc.2013.230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Goto Y., Kojima S., Nishikawa R., Kurozumi A., Kato M., Enokida H., Matsushita R., Yamazaki K., Ishida Y., Nakagawa M., Naya Y., Ichikawa T., Seki N. MicroRNA expression signature of castration-resistant prostate cancer: the microRNA-221/222 cluster functions as a tumour suppressor and disease progression marker. Br. J. Cancer. 2015;113:1055–1065. doi: 10.1038/bjc.2015.300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Li Y., Vandenboom T.G., II, Wang Z., Kong D., Ali S., Philip P.A., Sarkar F.H. miR-146a suppresses invasion of pancreatic cancer cells. Cancer Res. 2010;70:1486–1495. doi: 10.1158/0008-5472.CAN-09-2792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Chen G., Umelo I.A., Lv S., Teugels E., Fostier K., Kronenberger P., Dewaele A., Sadones J., Geers C., De Greve J. miR-146a inhibits cell growth, cell migration and induces apoptosis in non-small cell lung cancer cells. PLoS One. 2013;8:e60317. doi: 10.1371/journal.pone.0060317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lin S.L., Chiang A., Chang D., Ying S.Y. Loss of mir-146a function in hormone-refractory prostate cancer. RNA. 2008;14:417–424. doi: 10.1261/rna.874808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Vigil D., Kim T.Y., Plachco A., Garton A.J., Castaldo L., Pachter J.A., Dong H., Chen X., Tokar B., Campbell S.L., Der C.J. ROCK1 and ROCK2 are required for non-small cell lung cancer anchorage-independent growth and invasion. Cancer Res. 2012;72:5338–5347. doi: 10.1158/0008-5472.CAN-11-2373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Xu B., Huang Y., Niu X., Tao T., Jiang L., Tong N., Chen S., Liu N., Zhu W., Chen M. Hsa-miR-146a-5p modulates androgen-independent prostate cancer cells apoptosis by targeting ROCK1. Prostate. 2015;75:1896–1903. doi: 10.1002/pros.23068. [DOI] [PubMed] [Google Scholar]
- 49.Kobayashi T., Inoue T., Shimizu Y., Terada N., Maeno A., Kajita Y., Yamasaki T., Kamba T., Toda Y., Mikami Y., Yamada T., Kamoto T., Ogawa O., Nakamura E. Activation of Rac1 is closely related to androgen-independent cell proliferation of prostate cancer cells both in vitro and in vivo. Mol. Endocrinol. 2010;24:722–734. doi: 10.1210/me.2009-0326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sun Q., Zhao X., Liu X., Wang Y., Huang J., Jiang B., Chen Q., Yu J. miR-146a functions as a tumor suppressor in prostate cancer by targeting Rac1. Prostate. 2014;74:1613–1621. doi: 10.1002/pros.22878. [DOI] [PubMed] [Google Scholar]
- 51.Tannock I.F., de Wit R., Berry W.R., Horti J., Pluzanska A., Chi K.N., Oudard S., Theodore C., James N.D., Turesson I., Rosenthal M.A., Eisenberger M.A., Investigators T.A.X. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N. Engl. J. Med. 2004;351:1502–1512. doi: 10.1056/NEJMoa040720. [DOI] [PubMed] [Google Scholar]
- 52.Petrylak D.P., Tangen C.M., Hussain M.H., Lara P.N., Jr, Jones J.A., Taplin M.E., Burch P.A., Berry D., Moinpour C., Kohli M., Benson M.C., Small E.J., Raghavan D., Crawford E.D. Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N. Engl. J. Med. 2004;351:1513–1520. doi: 10.1056/NEJMoa041318. [DOI] [PubMed] [Google Scholar]
- 53.Stein C.A. Mechanisms of action of taxanes in prostate cancer. Semin. Oncol. 1999;26:3–7. [PubMed] [Google Scholar]
- 54.de Bono J.S., Oudard S., Ozguroglu M., Hansen S., Machiels J.P., Kocak I., Gravis G., Bodrogi I., Mackenzie M.J., Shen L., Roessner M., Gupta S., Sartor A.O., Investigators T. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet. 2010;376:1147–1154. doi: 10.1016/S0140-6736(10)61389-X. [DOI] [PubMed] [Google Scholar]
- 55.Bonci D., Coppola V., Musumeci M., Addario A., Giuffrida R., Memeo L., D’Urso L., Pagliuca A., Biffoni M., Labbaye C., Bartucci M., Muto G., Peschle C., De Maria R. The miR-15a-miR-16-1 cluster controls prostate cancer by targeting multiple oncogenic activities. Nat. Med. 2008;14:1271–1277. doi: 10.1038/nm.1880. [DOI] [PubMed] [Google Scholar]
- 56.Takeshita F., Patrawala L., Osaki M., Takahashi R.U., Yamamoto Y., Kosaka N., Kawamata M., Kelnar K., Bader A.G., Brown D., Ochiya T. Systemic delivery of synthetic microRNA-16 inhibits the growth of metastatic prostate tumors via downregulation of multiple cell-cycle genes. Mol. Ther. 2010;18:181–187. doi: 10.1038/mt.2009.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Hellwinkel O.J.C., Sellier C., Sylvester Y.J., Brase J.C., Isbarn H., Erbersdobler A., Steuber T., Sültmann H., Schlomm T., Wagner C. A Cancer-indicative microRNA pattern in normal prostate tissue. Int. J. Mol. Sci. 2013;14(3):5239–5249. doi: 10.3390/ijms14035239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Porkka K.P., Ogg E.L., Saramaki O.R., Vessella R.L., Pukkila H., Lahdesmaki H., van Weerden W.M., Wolf M., Kallioniemi O.P., Jenster G., Visakorpi T. The miR-15a-miR-16-1 locus is homozygously deleted in a subset of prostate cancers. Genes Chromosomes Cancer. 2011;50:499–509. doi: 10.1002/gcc.20873. [DOI] [PubMed] [Google Scholar]
- 59.Todorova K., Hayrabedyan S. Mir-15A reconstitution in prostate cancer cell line suppresses cancer progression through down regulation of MYB and androgen receptor upregulation. Acta Med. Bulg. 2015;42:18–22. [Google Scholar]
- 60.Bonci D., Coppola V., Patrizii M., Addario A., Cannistraci A., Francescangeli F., Pecci R., Muto G., Collura D., Bedini R., Zeuner A., Valtieri M., Sentinelli S., Benassi M.S., Gallucci M., Carlini P., Piccolo S., De Maria R. A microRNA code for prostate cancer metastasis. Oncogene. 2016;35:1180–1192. doi: 10.1038/onc.2015.176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Musumeci M., Coppola V., Addario A., Patrizii M., Maugeri-Sacca M., Memeo L., Colarossi C., Francescangeli F., Biffoni M., Collura D., Giacobbe A., D’Urso L., Falchi M., Venneri M.A., Muto G., De Maria R., Bonci D. Control of tumor and microenvironment cross-talk by miR-15a and miR-16 in prostate cancer. Oncogene. 2011;30:4231–4242. doi: 10.1038/onc.2011.140. [DOI] [PubMed] [Google Scholar]
- 62.Banyard J., Chung I., Wilson A.M., Vetter G., Le Bechec A., Bielenberg D.R., Zetter B.R. Regulation of epithelial plasticity by miR-424 and miR-200 in a new prostate cancer metastasis model. Sci. Rep. 2013;3:3151. doi: 10.1038/srep03151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Puhr M., Hoefer J., Schafer G., Erb H.H., Oh S.J., Klocker H., Heidegger I., Neuwirt H., Culig Z. Epithelial-to-mesenchymal transition leads to docetaxel resistance in prostate cancer and is mediated by reduced expression of miR-200c and miR-205. Am. J. Pathol. 2012;181:2188–2201. doi: 10.1016/j.ajpath.2012.08.011. [DOI] [PubMed] [Google Scholar]
- 64.Massoner P., Thomm T., Mack B., Untergasser G., Martowicz A., Bobowski K., Klocker H., Gires O., Puhr M. EpCAM is overexpressed in local and metastatic prostate cancer, suppressed by chemotherapy and modulated by MET-associated miRNA-200c/205. Br. J. Cancer. 2014;111:955–964. doi: 10.1038/bjc.2014.366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Shi R., Xiao H., Yang T., Chang L., Tian Y., Wu B., Xu H. Effects of miR-200c on the migration and invasion abilities of human prostate cancer Du145 cells and the corresponding mechanism. Front. Med. 2014;8:456–463. doi: 10.1007/s11684-014-0353-z. [DOI] [PubMed] [Google Scholar]
- 66.Herman J.G., Baylin S.B. Gene silencing in cancer in association with promoter hypermethylation. N. Engl. J. Med. 2003;349:2042–2054. doi: 10.1056/NEJMra023075. [DOI] [PubMed] [Google Scholar]
- 67.Vrba L., Jensen T.J., Garbe J.C., Heimark R.L., Cress A.E., Dickinson S., Stampfer M.R., Futscher B.W. Role for DNA methylation in the regulation of miR-200c and miR-141 expression in normal and cancer cells. PLoS One. 2010;5:e8697. doi: 10.1371/journal.pone.0008697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Gu Y., Lei D., Qin X., Chen P., Zou Y.M., Hu Y. Integrated analysis reveals together miR-182, miR-200c and miR-221 can help in the diagnosis of prostate cancer. PLoS One. 2015;10:e0140862. doi: 10.1371/journal.pone.0140862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Abdullah L.N., Chow E.K. Mechanisms of chemoresistance in cancer stem cells. Clin. Transl. Med. 2013;2:3. doi: 10.1186/2001-1326-2-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Singh S., Chitkara D., Mehrazin R., Behrman S.W., Wake R.W., Mahato R.I. Chemoresistance in prostate cancer cells is regulated by miRNAs and Hedgehog pathway. PLoS One. 2012;7:e40021. doi: 10.1371/journal.pone.0040021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Khan A.P., Poisson L.M., Bhat V.B., Fermin D., Zhao R., Kalyana-Sundaram S., Michailidis G., Nesvizhskii A.I., Omenn G.S., Chinnaiyan A.M., Sreekumar A. Quantitative proteomic profiling of prostate cancer reveals a role for miR-128 in prostate cancer. Mol. Cell. Proteomics. 2010;9:298–312. doi: 10.1074/mcp.M900159-MCP200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Sun X., Yang Z., Zhang Y., He J., Wang F., Su P., Han J., Song Z., Fei Y. Prognostic implications of tissue and serum levels of microRNA-128 in human prostate cancer. Int. J. Clin. Exp. Pathol. 2015;8:8394–8401. [PMC free article] [PubMed] [Google Scholar]
- 73.Jin M., Zhang T., Liu C., Badeaux M.A., Liu B., Liu R., Jeter C., Chen X., Vlassov A.V., Tang D.G. miRNA-128 suppresses prostate cancer by inhibiting BMI-1 to inhibit tumor-initiating cells. Cancer Res. 2014;74:4183–4195. doi: 10.1158/0008-5472.CAN-14-0404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Lukacs R.U., Memarzadeh S., Wu H., Witte O.N. Bmi-1 is a crucial regulator of prostate stem cell self-renewal and malignant transformation. Cell Stem Cell. 2010;7:682–693. doi: 10.1016/j.stem.2010.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Clape C., Fritz V., Henriquet C., Apparailly F., Fernandez P.L., Iborra F., Avances C., Villalba M., Culine S., Fajas L. miR-143 interferes with ERK5 signaling, and abrogates prostate cancer progression in mice. PLoS One. 2009;4:e7542. doi: 10.1371/journal.pone.0007542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zhou P., Chen W.G., Li X.W. MicroRNA-143 acts as a tumor suppressor by targeting hexokinase 2 in human prostate cancer. Am. J. Cancer Res. 2015;5:2056–2063. [PMC free article] [PubMed] [Google Scholar]
- 77.Peng X., Guo W., Liu T., Wang X., Tu X., Xiong D., Chen S., Lai Y., Du H., Chen G., Liu G., Tang Y., Huang S., Zou X. Identification of miRs-143 and -145 that is associated with bone metastasis of prostate cancer and involved in the regulation of EMT. PLoS One. 2011;6:e20341. doi: 10.1371/journal.pone.0020341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Fan X., Chen X., Deng W., Zhong G., Cai Q., Lin T. Up-regulated microRNA-143 in cancer stem cells differentiation promotes prostate cancer cells metastasis by modulating FNDC3B expression. BMC Cancer. 2013;13:61. doi: 10.1186/1471-2407-13-61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Weber M.J., Gioeli D. Ras signaling in prostate cancer progression. J. Cell. Biochem. 2004;91:13–25. doi: 10.1002/jcb.10683. [DOI] [PubMed] [Google Scholar]
- 80.Xu B., Niu X., Zhang X., Tao J., Wu D., Wang Z., Li P., Zhang W., Wu H., Feng N., Wang Z., Hua L., Wang X. miR-143 decreases prostate cancer cells proliferation and migration and enhances their sensitivity to docetaxel through suppression of KRAS. Mol. Cell. Biochem. 2011;350:207–213. doi: 10.1007/s11010-010-0700-6. [DOI] [PubMed] [Google Scholar]
- 81.Ahmad I., Singh L.B., Yang Z.H., Kalna G., Fleming J., Fisher G., Cooper C., Cuzick J., Berney D.M., Moller H., Scardino P., Leung H.Y. Mir143 expression inversely correlates with nuclear ERK5 immunoreactivity in clinical prostate cancer. Br. J. Cancer. 2013;108:149–154. doi: 10.1038/bjc.2012.510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Cai C., Rajaram M., Zhou X., Liu Q., Marchica J., Li J., Powers R.S. Activation of multiple cancer pathways and tumor maintenance function of the 3q amplified oncogene FNDC3B. Cell Cycle. 2012;11:1773–1781. doi: 10.4161/cc.20121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Cheung C.C., Chung G.T., Lun S.W., To K.F., Choy K.W., Lau K.M., Siu S.P., Guan X.Y., Ngan R.K., Yip T.T., Busson P., Tsao S.W., Lo K.W. miR-31 is consistently inactivated in EBV-associated nasopharyngeal carcinoma and contributes to its tumorigenesis. Mol. Cancer. 2014;13:184. doi: 10.1186/1476-4598-13-184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kim H.S., Lee K.S., Bae H.J., Eun J.W., Shen Q., Park S.J., Shin W.C., Yang H.D., Park M., Park W.S., Kang Y.K., Nam S.W. MicroRNA-31 functions as a tumor suppressor by regulating cell cycle and epithelial-mesenchymal transition regulatory proteins in liver cancer. Oncotarget. 2015;6:8089–8102. doi: 10.18632/oncotarget.3512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Ning Z., Zhu H., Li F., Liu Q., Liu G., Tan T., Zhang B., Chen S., Li G., Huang D., Meltzer S.J., Zhang H. Tumor suppression by miR-31 in esophageal carcinoma is p21-dependent. Genes Cancer. 2014;5:436–444. doi: 10.18632/genesandcancer.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Rasheed S.A., Teo C.R., Beillard E.J., Voorhoeve P.M., Zhou W., Ghosh S., Casey P.J. MicroRNA-31 controls G protein alpha-13 (GNA13) expression and cell invasion in breast cancer cells. Mol. Cancer. 2015;14:67. doi: 10.1186/s12943-015-0337-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Bhatnagar N., Li X., Padi S.K., Zhang Q., Tang M.S., Guo B. Downregulation of miR-205 and miR-31 confers resistance to chemotherapy-induced apoptosis in prostate cancer cells. Cell Death Dis. 2010;1:e105. doi: 10.1038/cddis.2010.85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Lin P.C., Chiu Y.L., Banerjee S., Park K., Mosquera J.M., Giannopoulou E., Alves P., Tewari A.K., Gerstein M.B., Beltran H., Melnick A.M., Elemento O., Demichelis F., Rubin M.A. Epigenetic repression of miR-31 disrupts androgen receptor homeostasis and contributes to prostate cancer progression. Cancer Res. 2013;73:1232–1244. doi: 10.1158/0008-5472.CAN-12-2968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Yang W.W., Shu B., Zhu Y., Yang H.T. E2F6 inhibits cobalt chloride-mimetic hypoxia-induced apoptosis through E2F1. Mol. Biol. Cell. 2008;19:3691–3700. doi: 10.1091/mbc.E08-02-0171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Yang W.W., Wang Z.H., Zhu Y., Yang H.T. E2F6 negatively regulates ultraviolet-induced apoptosis via modulation of BRCA1. Cell Death Differ. 2007;14:807–817. doi: 10.1038/sj.cdd.4402062. [DOI] [PubMed] [Google Scholar]
- 91.Zhang Q., Sun M., Zhou S., Guo B. Class I HDAC inhibitor mocetinostat induces apoptosis by activation of miR-31 expression and suppression of E2F6. Cell Death Discov. 2016;2:16036. doi: 10.1038/cddiscovery.2016.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Liu C., Kelnar K., Liu B., Chen X., Calhoun-Davis T., Li H., Patrawala L., Yan H., Jeter C., Honorio S., Wiggins J.F., Bader A.G., Fagin R., Brown D., Tang D.G. The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat. Med. 2011;17:211–215. doi: 10.1038/nm.2284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Liang J., Li Y., Daniels G., Sfanos K., De Marzo A., Wei J., Li X., Chen W., Wang J., Zhong X., Melamed J., Zhao J., Lee P. LEF1 Targeting EMT in prostate cancer invasion is regulated by miR-34a. Mol. Cancer Res. 2015;13:681–688. doi: 10.1158/1541-7786.MCR-14-0503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kojima K., Fujita Y., Nozawa Y., Deguchi T., Ito M. MiR-34a attenuates paclitaxel-resistance of hormone-refractory prostate cancer PC3 cells through direct and indirect mechanisms. Prostate. 2010;70:1501–1512. doi: 10.1002/pros.21185. [DOI] [PubMed] [Google Scholar]
- 95.Corcoran C., Rani S., O’Driscoll L. miR-34a is an intracellular and exosomal predictive biomarker for response to docetaxel with clinical relevance to prostate cancer progression. Prostate. 2014;74:1320–1334. doi: 10.1002/pros.22848. [DOI] [PubMed] [Google Scholar]
- 96.Yamamura S., Saini S., Majid S., Hirata H., Ueno K., Deng G., Dahiya R. MicroRNA-34a modulates c-Myc transcriptional complexes to suppress malignancy in human prostate cancer cells. PLoS One. 2012;7:e29722. doi: 10.1371/journal.pone.0029722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Chen W.Y., Liu S.Y., Chang Y.S., Yin J.J., Yeh H.L., Mouhieddine T.H., Hadadeh O., Abou-Kheir W., Liu Y.N. MicroRNA-34a regulates WNT/TCF7 signaling and inhibits bone metastasis in Ras-activated prostate cancer. Oncotarget. 2015;6:441–457. doi: 10.18632/oncotarget.2690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Gaur S., Wen Y., Song J.H., Parikh N.U., Mangala L.S., Blessing A.M., Ivan C., Wu S.Y., Varkaris A., Shi Y., Lopez-Berestein G., Frigo D.E., Sood A.K., Gallick G.E. Chitosan nanoparticle-mediated delivery of miRNA-34a decreases prostate tumor growth in the bone and its expression induces non-canonical autophagy. Oncotarget. 2015;6:29161–29177. doi: 10.18632/oncotarget.4971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Fujita Y., Kojima K., Hamada N., Ohhashi R., Akao Y., Nozawa Y., Deguchi T., Ito M. Effects of miR-34a on cell growth and chemoresistance in prostate cancer PC3 cells. Biochem. Biophys. Res. Commun. 2008;377:114–119. doi: 10.1016/j.bbrc.2008.09.086. [DOI] [PubMed] [Google Scholar]
- 100.Szostak M.J., Kyprianou N. Radiation-induced apoptosis: predictive and therapeutic significance in radiotherapy of prostate cancer. Oncol. Rep. 2000;7:699–706. doi: 10.3892/or.7.4.699. [review]. [DOI] [PubMed] [Google Scholar]
- 101.Kuban D.A., Thames H.D., Levy L.B., Horwitz E.M., Kupelian P.A., Martinez A.A., Michalski J.M., Pisansky T.M., Sandler H.M., Shipley W.U., Zelefsky M.J., Zietman A.L. Long-term multi-institutional analysis of stage T1-T2 prostate cancer treated with radiotherapy in the PSA era. Int. J. Radiat. Oncol. Biol. Phys. 2003;57:915–928. doi: 10.1016/s0360-3016(03)00632-1. [DOI] [PubMed] [Google Scholar]
- 102.Khuntia D., Reddy C.A., Mahadevan A., Klein E.A., Kupelian P.A. Recurrence-free survival rates after external-beam radiotherapy for patients with clinical T1-T3 prostate carcinoma in the prostate-specific antigen era: what should we expect? Cancer. 2004;100:1283–1292. doi: 10.1002/cncr.20093. [DOI] [PubMed] [Google Scholar]
- 103.Josson S., Sung S.Y., Lao K., Chung L.W., Johnstone P.A. Radiation modulation of microRNA in prostate cancer cell lines. Prostate. 2008;68:1599–1606. doi: 10.1002/pros.20827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Huang Z., Huang S., Wang Q., Liang L., Ni S., Wang L., Sheng W., He X., Du X. MicroRNA-95 promotes cell proliferation and targets sorting Nexin 1 in human colorectal carcinoma. Cancer Res. 2011;71:2582–2589. doi: 10.1158/0008-5472.CAN-10-3032. [DOI] [PubMed] [Google Scholar]
- 105.Chen X., Chen S., Hang W., Huang H., Ma H. MiR-95 induces proliferation and chemo- or radioresistance through directly targeting sorting nexin1 (SNX1) in non-small cell lung cancer. Biomed. Pharmacother. 2014;68:589–595. doi: 10.1016/j.biopha.2014.04.008. [DOI] [PubMed] [Google Scholar]
- 106.Huang X., Taeb S., Jahangiri S., Emmenegger U., Tran E., Bruce J., Mesci A., Korpela E., Vesprini D., Wong C.S., Bristow R.G., Liu F.F., Liu S.K. miRNA-95 mediates radioresistance in tumors by targeting the sphingolipid phosphatase SGPP1. Cancer Res. 2013;73:6972–6986. doi: 10.1158/0008-5472.CAN-13-1657. [DOI] [PubMed] [Google Scholar]
- 107.Gao X.Y., Li L., Wang X.H., Wen X.Z., Ji K., Ye L., Cai J., Jiang W.G., Ji J.F. Inhibition of sphingosine-1-phosphate phosphatase 1 promotes cancer cells migration in gastric cancer: clinical implications. Oncol. Rep. 2015;34:1977–1987. doi: 10.3892/or.2015.4162. [DOI] [PubMed] [Google Scholar]
- 108.Yau W.L., Lam C.S., Ng L., Chow A.K., Chan S.T., Chan J.Y., Wo J.Y., Ng K.T., Man K., Poon R.T., Pang R.W. Over-expression of miR-106b promotes cell migration and metastasis in hepatocellular carcinoma by activating epithelial-mesenchymal transition process. PLoS One. 2013;8:e57882. doi: 10.1371/journal.pone.0057882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Cheng Y., Guo Y., Zhang Y., You K., Li Z., Geng L. MicroRNA-106b is involved in transforming growth factor beta1-induced cell migration by targeting disabled homolog 2 in cervical carcinoma. J. Exp. Clin. Cancer Res. 2016;35:11. doi: 10.1186/s13046-016-0290-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Zhang G.J., Li J.S., Zhou H., Xiao H.X., Li Y., Zhou T. MicroRNA-106b promotes colorectal cancer cell migration and invasion by directly targeting DLC1. J. Exp. Clin. Cancer Res. 2015;34:73. doi: 10.1186/s13046-015-0189-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Hudson R.S., Yi M., Esposito D., Glynn S.A., Starks A.M., Yang Y., Schetter A.J., Watkins S.K., Hurwitz A.A., Dorsey T.H., Stephens R.M., Croce C.M., Ambs S. MicroRNA-106b-25 cluster expression is associated with early disease recurrence and targets caspase-7 and focal adhesion in human prostate cancer. Oncogene. 2013;32:4139–4147. doi: 10.1038/onc.2012.424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Li B., Shi X.B., Nori D., Chao C.K., Chen A.M., Valicenti R., White Rde V. Down-regulation of microRNA 106b is involved in p21-mediated cell cycle arrest in response to radiation in prostate cancer cells. Prostate. 2011;71:567–574. doi: 10.1002/pros.21272. [DOI] [PubMed] [Google Scholar]
- 113.Cheng C., Chen Z.Q., Shi X.T. MicroRNA-320 inhibits osteosarcoma cells proliferation by directly targeting fatty acid synthase. Tumour Biol. 2014;35:4177–4183. doi: 10.1007/s13277-013-1546-9. [DOI] [PubMed] [Google Scholar]
- 114.Sun J.Y., Xiao W.Z., Wang F., Wang Y.Q., Zhu Y.H., Wu Y.F., Miao Z.L., Lin Y.C. MicroRNA-320 inhibits cell proliferation in glioma by targeting E2F1. Mol. Med. Rep. 2015;12:2355–2359. doi: 10.3892/mmr.2015.3657. [DOI] [PubMed] [Google Scholar]
- 115.Zhang T., Zou P., Wang T., Xiang J., Cheng J., Chen D., Zhou J. Down-regulation of miR-320 associated with cancer progression and cell apoptosis via targeting Mcl-1 in cervical cancer. Tumour Biol. 2016;37:8931–8940. doi: 10.1007/s13277-015-4771-6. [DOI] [PubMed] [Google Scholar]
- 116.Hsieh I.S., Chang K.C., Tsai Y.T., Ke J.Y., Lu P.J., Lee K.H., Yeh S.D., Hong T.M., Chen Y.L. MicroRNA-320 suppresses the stem cell-like characteristics of prostate cancer cells by downregulating the Wnt/beta-catenin signaling pathway. Carcinogenesis. 2013;34:530–538. doi: 10.1093/carcin/bgs371. [DOI] [PubMed] [Google Scholar]
- 117.Okato A., Goto Y., Kurozumi A., Kato M., Kojima S., Matsushita R., Yonemori M., Miyamoto K., Ichikawa T., Seki N. Direct regulation of LAMP1 by tumor-suppressive microRNA-320a in prostate cancer. Int. J. Oncol. 2016;49:111–122. doi: 10.3892/ijo.2016.3522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Sato S., Katsushima K., Shinjo K., Hatanaka A., Ohka F., Suzuki S., Naiki-Ito A., Soga N., Takahashi S., Kondo Y. Histone deacetylase inhibition in prostate cancer triggers miR-320-mediated suppression of the androgen receptor. Cancer Res. 2016 doi: 10.1158/0008-5472.CAN-15-3339. [DOI] [PubMed] [Google Scholar]
- 119.Sarafian V., Jadot M., Foidart J.M., Letesson J.J., Van den Brule F., Castronovo V., Wattiaux R., Coninck S.W. Expression of Lamp-1 and Lamp-2 and their interactions with galectin-3 in human tumor cells. Int. J. Cancer. 1998;75:105–111. doi: 10.1002/(sici)1097-0215(19980105)75:1<105::aid-ijc16>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
- 120.Valkenburg K.C., Graveel C.R., Zylstra-Diegel C.R., Zhong Z., Williams B.O. Wnt/beta-catenin signaling in normal and cancer stem cells. Cancers (Basel) 2011;3:2050–2079. doi: 10.3390/cancers3022050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Feng Y.H., Tsao C.J. Emerging role of microRNA-21 in cancer. Biomed. Rep. 2016;5:395–402. doi: 10.3892/br.2016.747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Liu L.Z., Li C., Chen Q., Jing Y., Carpenter R., Jiang Y., Kung H.F., Lai L., Jiang B.H. MiR-21 induced angiogenesis through AKT and ERK activation and HIF-1alpha expression. PLoS One. 2011;6:e19139. doi: 10.1371/journal.pone.0019139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Amankwah E.K., Anegbe E., Park H., Pow-Sang J., Hakam A., Park J.Y. miR-21, miR-221 and miR-222 expression and prostate cancer recurrence among obese and non-obese cases. Asian J. Androl. 2013;15:226–230. doi: 10.1038/aja.2012.160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Ribas J., Ni X., Haffner M., Wentzel E.A., Salmasi A.H., Chowdhury W.H., Kudrolli T.A., Yegnasubramanian S., Luo J., Rodriguez R., Mendell J.T., Lupold S.E. miR-21: an androgen receptor-regulated microRNA that promotes hormone-dependent and hormone-independent prostate cancer growth. Cancer Res. 2009;69:7165–7169. doi: 10.1158/0008-5472.CAN-09-1448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Shi G.H., Ye D.W., Yao X.D., Zhang S.L., Dai B., Zhang H.L., Shen Y.J., Zhu Y., Zhu Y.P., Xiao W.J., Ma C.G. Involvement of microRNA-21 in mediating chemo-resistance to docetaxel in androgen-independent prostate cancer PC3 cells. Acta Pharmacol. Sin. 2010;31:867–873. doi: 10.1038/aps.2010.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Lu Z., Liu M., Stribinskis V., Klinge C.M., Ramos K.S., Colburn N.H., Li Y. MicroRNA-21 promotes cell transformation by targeting the programmed cell death 4 gene. Oncogene. 2008;27:4373–4379. doi: 10.1038/onc.2008.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Asangani I.A., Rasheed S.A., Nikolova D.A., Leupold J.H., Colburn N.H., Post S., Allgayer H. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene. 2008;27:2128–2136. doi: 10.1038/sj.onc.1210856. [DOI] [PubMed] [Google Scholar]
- 128.Dong B., Shi Z., Wang J., Wu J., Yang Z., Fang K. IL-6 Inhibits the targeted modulation of PDCD4 by miR-21 in prostate cancer. PLoS One. 2015;10:e0134366. doi: 10.1371/journal.pone.0134366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Reis S.T., Pontes-Junior J., Antunes A.A., Dall’Oglio M.F., Dip N., Passerotti C.C., Rossini G.A., Morais D.R., Nesrallah A.J., Piantino C., Srougi M., Leite K.R. miR-21 may acts as an oncomir by targeting RECK, a matrix metalloproteinase regulator, in prostate cancer. BMC Urol. 2012;12:14. doi: 10.1186/1471-2490-12-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Leite K.R., Reis S.T., Viana N., Morais D.R., Moura C.M., Silva I.A., Pontes J., Jr, Katz B., Srougi M. Controlling RECK miR21 promotes tumor cell invasion and is related to biochemical recurrence in prostate cancer. J. Cancer. 2015;6:292–301. doi: 10.7150/jca.11038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Mishra S., Lin C.L., Huang T.H., Bouamar H., Sun L.Z. MicroRNA-21 inhibits p57Kip2 expression in prostate cancer. Mol. Cancer. 2014;13:212. doi: 10.1186/1476-4598-13-212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Zhu D., Chen H., Yang X., Chen W., Wang L., Xu J., Yu L. miR-32 functions as a tumor suppressor and directly targets SOX9 in human non-small cell lung cancer. OncoTargets Ther. 2015;8:1773–1783. doi: 10.2147/OTT.S72457. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 133.Li L., Wu D. miR-32 inhibits proliferation, epithelial-mesenchymal transition, and metastasis by targeting TWIST1 in non-small-cell lung cancer cells. OncoTargets Ther. 2016;9:1489–1498. doi: 10.2147/OTT.S99931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Zhang J., Kuai X., Song M., Chen X., Yu Z., Zhang H., Mao Z. microRNA-32 inhibits the proliferation and invasion of the SGC-7901 gastric cancer cell line in vitro. Oncol. Lett. 2014;7:270–274. doi: 10.3892/ol.2013.1667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Wu W., Yang J., Feng X., Wang H., Ye S., Yang P., Tan W., Wei G., Zhou Y. MicroRNA-32 (miR-32) regulates phosphatase and tensin homologue (PTEN) expression and promotes growth, migration, and invasion in colorectal carcinoma cells. Mol. Cancer. 2013;12:30. doi: 10.1186/1476-4598-12-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Jalava S.E., Urbanucci A., Latonen L., Waltering K.K., Sahu B., Janne O.A., Seppala J., Lahdesmaki H., Tammela T.L., Visakorpi T. Androgen-regulated miR-32 targets BTG2 and is overexpressed in castration-resistant prostate cancer. Oncogene. 2012;31:4460–4471. doi: 10.1038/onc.2011.624. [DOI] [PubMed] [Google Scholar]
- 137.Coppola V., Musumeci M., Patrizii M., Cannistraci A., Addario A., Maugeri-Sacca M., Biffoni M., Francescangeli F., Cordenonsi M., Piccolo S., Memeo L., Pagliuca A., Muto G., Zeuner A., De Maria R., Bonci D. BTG2 loss and miR-21 upregulation contribute to prostate cell transformation by inducing luminal markers expression and epithelial-mesenchymal transition. Oncogene. 2013;32:1843–1853. doi: 10.1038/onc.2012.194. [DOI] [PubMed] [Google Scholar]
- 138.Liao H., Xiao Y., Hu Y., Xiao Y., Yin Z., Liu L. microRNA-32 induces radioresistance by targeting DAB2IP and regulating autophagy in prostate cancer cells. Oncol. Lett. 2015;10:2055–2062. doi: 10.3892/ol.2015.3551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Xie D., Gore C., Liu J., Pong R.C., Mason R., Hao G., Long M., Kabbani W., Yu L., Zhang H., Chen H., Sun X., Boothman D.A., Min W., Hsieh J.T. Role of DAB2IP in modulating epithelial-to-mesenchymal transition and prostate cancer metastasis. Proc. Natl. Acad. Sci. USA. 2010;107:2485–2490. doi: 10.1073/pnas.0908133107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Wang N., Li Q., Feng N.H., Cheng G., Guan Z.L., Wang Y., Qin C., Yin C.J., Hua L.X. miR-205 is frequently downregulated in prostate cancer and acts as a tumor suppressor by inhibiting tumor growth. Asian J. Androl. 2013;15:735–741. doi: 10.1038/aja.2013.80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Verdoodt B., Neid M., Vogt M., Kuhn V., Liffers S.T., Palisaar R.J., Noldus J., Tannapfel A., Mirmohammadsadegh A. MicroRNA-205, a novel regulator of the anti-apoptotic protein Bcl2, is downregulated in prostate cancer. Int. J. Oncol. 2013;43:307–314. doi: 10.3892/ijo.2013.1915. [DOI] [PubMed] [Google Scholar]
- 142.Majid S., Dar A.A., Saini S., Yamamura S., Hirata H., Tanaka Y., Deng G., Dahiya R. MicroRNA-205-directed transcriptional activation of tumor suppressor gen.es in prostate cancer. Cancer. 2010;116:5637–5649. doi: 10.1002/cncr.25488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Hagman Z., Haflidadottir B.S., Ceder J.A., Larne O., Bjartell A., Lilja H., Edsjo A., Ceder Y. miR-205 negatively regulates the androgen receptor and is associated with adverse outcome of prostate cancer patients. Br. J. Cancer. 2013;108:1668–1676. doi: 10.1038/bjc.2013.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Kalogirou C., Spahn M., Krebs M., Joniau S., Lerut E., Burger M., Scholz C.J., Kneitz S., Riedmiller H., Kneitz B. MiR-205 is progressively down-regulated in lymph node metastasis but fails as a prognostic biomarker in high-risk prostate cancer. Int. J. Mol. Sci. 2013;14:21414–21434. doi: 10.3390/ijms141121414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Hulf T., Sibbritt T., Wiklund E.D., Patterson K., Song J.Z., Stirzaker C., Qu W., Nair S., Horvath L.G., Armstrong N.J., Kench J.G., Sutherland R.L., Clark S.J. Epigenetic-induced repression of microRNA-205 is associated with MED1 activation and a poorer prognosis in localized prostate cancer. Oncogene. 2013;32:2891–2899. doi: 10.1038/onc.2012.300. [DOI] [PubMed] [Google Scholar]
- 146.Tucci P., Agostini M., Grespi F., Markert E.K., Terrinoni A., Vousden K.H., Muller P.A., Dotsch V., Kehrloesser S., Sayan B.S., Giaccone G., Lowe S.W., Takahashi N., Vandenabeele P., Knight R.A., Levine A.J., Melino G. Loss of p63 and its microRNA-205 target results in enhanced cell migration and metastasis in prostate cancer. Proc. Natl. Acad. Sci. USA. 2012;109:15312–15317. doi: 10.1073/pnas.1110977109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Wang W., Liu J., Wu Q. MiR-205 suppresses autophagy and enhances radiosensitivity of prostate cancer cells by targeting TP53INP1. Eur. Rev. Med. Pharmacol. Sci. 2016;20:92–100. [PubMed] [Google Scholar]
- 148.Xu C.G., Yang M.F., Fan J.X., Wang W. MiR-30a and miR-205 are downregulated in hypoxia and modulate radiosensitivity of prostate cancer cells by inhibiting autophagy via TP53INP1. Eur. Rev. Med. Pharmacol. Sci. 2016;20:1501–1508. [PubMed] [Google Scholar]
- 149.Lawrie C.H., Gal S., Dunlop H.M., Pushkaran B., Liggins A.P., Pulford K., Banham A.H., Pezzella F., Boultwood J., Wainscoat J.S., Hatton C.S., Harris A.L. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Br. J. Haematol. 2008;141:672–675. doi: 10.1111/j.1365-2141.2008.07077.x. [DOI] [PubMed] [Google Scholar]
- 150.Zernecke A., Bidzhekov K., Noels H., Shagdarsuren E., Gan L., Denecke B., Hristov M., Koppel T., Jahantigh M.N., Lutgens E., Wang S., Olson E.N., Schober A., Weber C. Delivery of microRNA-126 by apoptotic bodies induces CXCL12-dependent vascular protection. Sci. Signal. 2009;2:ra81. doi: 10.1126/scisignal.2000610. [DOI] [PubMed] [Google Scholar]
- 151.Valadi H., Ekstrom K., Bossios A., Sjostrand M., Lee J.J., Lotvall J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007;9:654–659. doi: 10.1038/ncb1596. [DOI] [PubMed] [Google Scholar]
- 152.Arroyo J.D., Chevillet J.R., Kroh E.M., Ruf I.K., Pritchard C.C., Gibson D.F., Mitchell P.S., Bennett C.F., Pogosova-Agadjanyan E.L., Stirewalt D.L., Tait J.F., Tewari M. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc. Natl. Acad. Sci. USA. 2011;108:5003–5008. doi: 10.1073/pnas.1019055108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Yaman Agaoglu F., Kovancilar M., Dizdar Y., Darendeliler E., Holdenrieder S., Dalay N., Gezer U. Investigation of miR-21, miR-141, and miR-221 in blood circulation of patients with prostate cancer. Tumour Biol. 2011;32:583–588. doi: 10.1007/s13277-011-0154-9. [DOI] [PubMed] [Google Scholar]
- 154.Gonzales J.C., Fink L.M., Goodman O.B., Jr, Symanowski J.T., Vogelzang N.J., Ward D.C. 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. Clin. Genitourin. Cancer. 2011;9:39–45. doi: 10.1016/j.clgc.2011.05.008. [DOI] [PubMed] [Google Scholar]
- 155.Bryant R.J., Pawlowski T., Catto J.W., Marsden G., Vessella R.L., Rhees B., Kuslich C., Visakorpi T., Hamdy F.C. Changes in circulating microRNA levels associated with prostate cancer. Br. J. Cancer. 2012;106:768–774. doi: 10.1038/bjc.2011.595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Chen Z.H., Zhang G.L., Li H.R., Luo J.D., Li Z.X., Chen G.M., Yang J. A panel of five circulating microRNAs as potential biomarkers for prostate cancer. Prostate. 2012;72:1443–1452. doi: 10.1002/pros.22495. [DOI] [PubMed] [Google Scholar]
- 157.Shen J., Hruby G.W., McKiernan J.M., Gurvich I., Lipsky M.J., Benson M.C., Santella R.M. Dysregulation of circulating microRNAs and prediction of aggressive prostate cancer. Prostate. 2012;72:1469–1477. doi: 10.1002/pros.22499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Watahiki A., Macfarlane R.J., Gleave M.E., Crea F., Wang Y., Helgason C.D., Chi K.N. Plasma miRNAs as biomarkers to identify patients with castration-resistant metastatic prostate cancer. Int. J. Mol. Sci. 2013;14:7757–7770. doi: 10.3390/ijms14047757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Huang X., Yuan T., Liang M., Du M., Xia S., Dittmar R., Wang D., See W., Costello B.A., Quevedo F., Tan W., Nandy D., Bevan G.H., Longenbach S., Sun Z., Lu Y., Wang T., Thibodeau S.N., Boardman L., Kohli M., Wang L. Exosomal miR-1290 and miR-375 as prognostic markers in castration-resistant prostate cancer. Eur. Urol. 2015;67:33–41. doi: 10.1016/j.eururo.2014.07.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Lin H.M., Castillo L., Mahon K.L., Chiam K., Lee B.Y., Nguyen Q., Boyer M.J., Stockler M.R., Pavlakis N., Marx G., Mallesara G., Gurney H., Clark S.J., Swarbrick A., Daly R.J., Horvath L.G. Circulating microRNAs are associated with docetaxel chemotherapy outcome in castration-resistant prostate cancer. Br. J. Cancer. 2014;110:2462–2471. doi: 10.1038/bjc.2014.181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Lodes M.J., Caraballo M., Suciu D., Munro S., Kumar A., Anderson B. Detection of cancer with serum miRNAs on an oligonucleotide microarray. PLoS One. 2009;4:e6229. doi: 10.1371/journal.pone.0006229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Mahn R., Heukamp L.C., Rogenhofer S., von Ruecker A., Muller S.C., Ellinger J. Circulating microRNAs (miRNA) in serum of patients with prostate cancer. Urology. 2011;77(1265):e1269–e1216. doi: 10.1016/j.urology.2011.01.020. [DOI] [PubMed] [Google Scholar]
- 163.Moltzahn F., Olshen A.B., Baehner L., Peek A., Fong L., Stoppler H., Simko J., Hilton J.F., Carroll P., Blelloch R. Microfluidic-based multiplex qRT-PCR identifies diagnostic and prognostic microRNA signatures in the sera of prostate cancer patients. Cancer Res. 2011;71:550–560. doi: 10.1158/0008-5472.CAN-10-1229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Haldrup C., Kosaka N., Ochiya T., Borre M., Hoyer S., Orntoft T.F., Sorensen K.D. Profiling of circulating microRNAs for prostate cancer biomarker discovery. Drug Deliv. Transl. Res. 2014;4:19–30. doi: 10.1007/s13346-013-0169-4. [DOI] [PubMed] [Google Scholar]
- 165.Brase J.C., Johannes M., Schlomm T., Falth M., Haese A., Steuber T., Beissbarth T., Kuner R., Sultmann H. Circulating miRNAs are correlated with tumor progression in prostate cancer. Int. J. Cancer. 2011;128:608–616. doi: 10.1002/ijc.25376. [DOI] [PubMed] [Google Scholar]
- 166.Zhang H.L., Yang L.F., Zhu Y., Yao X.D., Zhang S.L., Dai B., Zhu Y.P., Shen Y.J., Shi G.H., Ye D.W. Serum miRNA-21: elevated levels in patients with metastatic hormone-refractory prostate cancer and potential predictive factor for the efficacy of docetaxel-based chemotherapy. Prostate. 2011;71:326–331. doi: 10.1002/pros.21246. [DOI] [PubMed] [Google Scholar]
- 167.Cheng H.H., Mitchell P.S., Kroh E.M., Dowell A.E., Chery L., Siddiqui J., Nelson P.S., Vessella R.L., Knudsen B.S., Chinnaiyan A.M., Pienta K.J., Morrissey C., Tewari M. Circulating microRNA profiling identifies a subset of metastatic prostate cancer patients with evidence of cancer-associated hypoxia. PLoS One. 2013;8:e69239. doi: 10.1371/journal.pone.0069239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Nguyen H.C., Xie W., Yang M., Hsieh C.L., Drouin S., Lee G.S., Kantoff P.W. Expression differences of circulating microRNAs in metastatic castration resistant prostate cancer and low-risk, localized prostate cancer. Prostate. 2013;73:346–354. doi: 10.1002/pros.22572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Stephenson A.J., Kattan M.W., Eastham J.A., Dotan Z.A., Bianco F.J., Jr, Lilja H., Scardino P.T. Defining biochemical recurrence of prostate cancer after radical prostatectomy: a proposal for a standardized definition. J. Clin. Oncol. 2006;24:3973–3978. doi: 10.1200/JCO.2005.04.0756. [DOI] [PubMed] [Google Scholar]
- 170.Selth L.A., Townley S.L., Bert A.G., Stricker P.D., Sutherland P.D., Horvath L.G., Goodall G.J., Butler L.M., Tilley W.D. Circulating microRNAs predict biochemical recurrence in prostate cancer patients. Br. J. Cancer. 2013;109:641–650. doi: 10.1038/bjc.2013.369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Mihelich B.L., Maranville J.C., Nolley R., Peehl D.M., Nonn L. Elevated serum microRNA levels associate with absence of high-grade prostate cancer in a retrospective cohort. PLoS One. 2015;10:e0124245. doi: 10.1371/journal.pone.0124245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Srivastava A., Goldberger H., Dimtchev A., Ramalinga M., Chijioke J., Marian C., Oermann E.K., Uhm S., Kim J.S., Chen L.N., Li X., Berry D.L., Kallakury B.V., Chauhan S.C., Collins S.P., Suy S., Kumar D. MicroRNA profiling in prostate cancer--the diagnostic potential of urinary miR-205 and miR-214. PLoS One. 2013;8:e76994. doi: 10.1371/journal.pone.0076994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Stephan C., Jung M., Rabenhorst S., Kilic E., Jung K. Urinary miR-183 and miR-205 do not surpass PCA3 in urine as predictive markers for prostate biopsy outcome despite their highly dysregulated expression in prostate cancer tissue. Clin. Chem. Lab. Med. 2015;53:1109–1118. doi: 10.1515/cclm-2014-1000. [DOI] [PubMed] [Google Scholar]
- 174.Haj-Ahmad T.A., Abdalla M.A., Haj-Ahmad Y. Potential urinary miRNA biomarker candidates for the accurate detection of prostate cancer among benign prostatic hyperplasia patients. J. Cancer. 2014;5:182–191. doi: 10.7150/jca.6799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Casanova-Salas I., Rubio-Briones J., Calatrava A., Mancarella C., Masia E., Casanova J., Fernandez-Serra A., Rubio L., Ramirez-Backhaus M., Arminan A., Dominguez-Escrig J., Martinez F., Garcia-Casado Z., Scotlandi K., Vicent M.J., Lopez-Guerrero J.A. Identification of miR-187 and miR-182 as biomarkers of early diagnosis and prognosis in patients with prostate cancer treated with radical prostatectomy. J. Urol. 2014;192:252–259. doi: 10.1016/j.juro.2014.01.107. [DOI] [PubMed] [Google Scholar]
- 176.Korzeniewski N., Tosev G., Pahernik S., Hadaschik B., Hohenfellner M., Duensing S. Identification of cell-free microRNAs in the urine of patients with prostate cancer. 2015. [DOI] [PubMed]
- 177.Selth L.A., Roberts M.J., Chow C.W., Marshall V.R., Doi S.A., Vincent A.D., Butler L.M., Lavin M.F., Tilley W.D., Gardiner R.A. Human seminal fluid as a source of prostate cancer-specific microRNA biomarkers. Endocr. Relat. Cancer. 2014;21:L17–L21. doi: 10.1530/ERC-14-0234. [DOI] [PubMed] [Google Scholar]
- 178.Roberts M.J., Chow C.W., Schirra H.J., Richards R., Buck M., Selth L.A., Doi S.A., Samaratunga H., Perry-Keene J., Payton D., Yaxley J., Lavin M.F., Gardiner R.A. Diagnostic performance of expression of PCA3, Hepsin and miR biomarkers inejaculate in combination with serum PSA for the detection of prostate cancer. Prostate. 2015;75:539–549. doi: 10.1002/pros.22942. [DOI] [PubMed] [Google Scholar]



