Skip to main content

Role of microRNAs in regulation of WNT signaling pathway in urothelial and prostate cancers

Abstract

Background

Urothelial cancer (UC) and prostate cancer (PCa) are the most common cancers among men with a high ratio of mortality in advanced-stages. The higher risk of these malignancies among men can be associated with higher carcinogens exposure. Molecular pathology of UC and PCa is related to the specific mutations and aberrations in some signaling pathways. WNT signaling is a highly regulated pathway that has a pivotal role during urothelial and prostate development and homeostasis. This pathway also plays a vital role in adult stem cell niches to maintain a balance between stemness and differentiation. Deregulation of the WNT pathway is frequently correlated with tumor progression and metastasis in urothelial and prostate tumors. Therefore, regulatory factors of WNT pathways are being investigated as diagnostic or prognostic markers and novel therapeutic targets during urothelial and prostate tumorigenesis. MicroRNAs (miRNAs) have a pivotal role in WNT signaling regulation in which there are interactions between miRNAs and WNT signaling pathway during tumor progression. Since, the miRNAs are sensitive, specific, and noninvasive, they can be introduced as efficient biomarkers of tumor progression.

Main body

In present review, we have summarized all of the miRNAs that have been involved in regulation of WNT signaling pathway in urothelial and prostate cancers.

Conclusions

It was observed that miRNAs were mainly involved in regulation of WNT signaling in bladder cancer cells through targeting the WNT ligands and cytoplasmic WNT components such as WNT5A, WNT7A, CTNNB1, GSK3β, and AXIN. Whereas, miRNAs were mainly involved in regulation of WNT signaling in prostate tumor cells via targeting the cytoplasmic WNT components and WNT related transcription factors such as CTNNB1, GSK3β, AXIN, TCF7, and LEF1. MiRNAs mainly functioned as tumor suppressors in bladder and prostate cancers through the WNT signaling inhibition. This review paves the way of introducing a noninvasive diagnostic panel of WNT related miRNAs in urothelial and prostate tumors.

Background

Urothelial cancer (UC) is the fourth most common cancer in men and the 10th cause of cancer related deaths worldwide with a high ratio of mortality in advanced-stage disease [1]. The higher risk of these malignancies among men can be associated with higher exposure to carcinogens such as smoking and occupation [2]. There are various UC environmental risk factors such as smoking, chronic inflammation of urinary tract, analgesics abuse, arylamines, arsenic contamination, radiotherapy, and cyclophosphamide [3]. Prostate cancer (PCa) is one of the most common neoplasia in males that ranks as the fifth leading cause of cancer mortality worldwide [4]. There are significant higher rates of PCa incidence and mortality in industrialized countries compared with developing countries that can be associated with diet, age, ethnicity, genetic factors, and family history [5]. Moreover, some factors are correlated with a high risk of PCa death including body mass index and blood pressure [4, 6]. PCa is a very heterogeneous disease with different clinical manifestations that is associated with a wide range of mutations and genetic abnormalities. PCa progression involves several stages in which mutations in prostate epithelial cells result in apoptosis suppression. Finally, the PCa cells obtain the castration-resistant phenotype with poor prognosis [7]. Although, surgical resection and radiotherapy are the common therapeutic methods for localized prostate tumor, androgen inhibition is the main therapeutic method for the tumor relapse [8]. However, majority of PCa patients progress toward the metastatic castration-resistant [9]. The standard treatment method for metastatic prostate cancer (mPCa) is androgen deprivation therapy (ADT) with or without chemotherapy. Radical prostatectomy (RP) and radiation therapy (RT) can also repress the metastatic tumors and improve the survival rates in mPCa patients [10]. RP and pelvic lymphadenectomy (PLDN) are the surgical methods that are principally proposed for advanced-stage prostate tumors. PLND is the most important strategy to identify the lymph node metastases in PCa. RT is also considered as the second key therapeutic strategy for high-risk prostate tumors [11]. Molecular pathology of PCa is related to the critical mutations in some signaling pathway molecules that affect or disrupt a cross-talk along the pathways. Bladder cancer (BCa) is the 10th most frequent cancer and the 13th leading cause of death worldwide [12, 13]. About 90% of BCa are identified as urothelial carcinoma of the bladder (UCB) [14]. The main risk factors of BCa are aromatic amines exposure, smoking, genetic and hormonal conditions, Schistosoma haematobium infection, and arsenic contamination of drinking water [15,16,17]. Sex hormone receptors, including androgen receptors (AR) and estrogen receptors (ERs), have been considered as critical factors for the sex differences in BCa. Accumulating evidences revealed that steroid hormone receptor signaling plays a key role in BCa progression [18]. Androgen receptors such as estrogen receptor-α and β promote tumor growth and chemo resistance in BCa [19,20,21]. UCB is also known as a highly heterogeneous malignancy with different histological subtypes, including non-muscle invasive bladder cancer (NMIBC) and muscle-invasive bladder cancer (MIBC) [22, 23]. About 90% of urothelial tumors are characterized as transitional cell carcinoma [24]. Urothelial tumors have also different differentiation patterns such as squamous, micropapillary, glandular, and clear cell [25]. Squamous type is the most common type with up to 40% of urothelial cancers (UC) [26]. Micropapillary urothelial carcinoma (MPUC) is an aggressive histopathological type that accounts 2–5% of UCs [27]. A combination therapy of surgical resection and chemo radiotherapy has been suggested for the BCa; however, there is a low 5-year survival rate in advanced BC patients [28]. Endoscopic resection is a routine treatment option [29]. Although, radical cystectomy or chemo radiotherapy are also common therapeutic methods for the muscle-invasive urothelial carcinoma (MIUC) cases, half of the cases progress toward relapse [30, 31].

WNT signaling is a highly regulated pathway that has a pivotal role during urothelial development and homeostasis [32]. This pathway also plays a vital role in adult stem cell niches to maintain a balance between self-renewal (undifferentiation) and differentiation [33]. WNT signaling is triggered through an interaction between WNT ligands and cysteine-rich G protein-coupled receptors (Frizzleds) and their associated co-receptors which results in activation of non-canonical and canonical pathways [34,35,36]. The presence of Wnt ligand leads to cytoplasmic accumulation of β-catenin that finally enters into the nucleus to regulate the WNT target genes as a transcriptional co-activator of T cell-specific factor (TCF)/lymphoid enhancer-binding factor (LEF) transcription factors [37, 38]. WNT signals are associated with epithelial-mesenchymal transition (EMT) regulators such as Snail, Twist, and Zeb [39, 40]. Deregulation of the WNT pathway is frequently correlated with tumor progression and metastasis in PCa [41, 42]. Cancer stem cells (CSCs) are known as tumor-initiating cells that are contributed with tumor metastasis [43, 44]. They have a high capacity of self-renewal and chemo-radiotherapeutic resistance by the developmental signaling pathways, EMT process, multi-drug resistance (MDR), and epigenetic changes [45, 46]. Genetic and epigenetic modifications in the regulatory components of WNT pathway result in acquiring urothelial CSC phenotype, chemo resistance, and decreased survival. Therefore, regulatory factors of WNT pathways are being investigated as diagnostic or prognostic markers and novel therapeutic targets during urothelial tumorigenesis [14]. MicroRNAs (miRNAs) are a class of non-coding RNAs which can directly interact with the 3′untranslated region (3′ UTR) of target mRNAs to induce their degradation or translation inhibition. They have a pivotal role in WNT signaling regulation. A network of miRNAs and WNT signaling pathway factors are involved in tumor progression of brain, colorectal, breast, liver, prostate, and other types of cancers [47]. EMT is a developmental process from an epithelial to invasive mesenchymal phenotype that has key roles in primary steps of tumor metastasis [48, 49]. There are many factors and signaling pathways associated with EMT including cadherins, vimentin, TGFb, and WNT signaling pathway [50]. EMT is associated with tumor metastasis via the WNT signaling pathway and miRNAs [51]. Various miRNAs such as miR-214, miR-320, miR-101, miR-1826, miR-548b, and miR-33a have been reported to suppress the WNT pathway by regulating β-catenin in different tumors [52]. Therefore, miRNAs can be used as therapeutic agents for the targeting of canonical WNT signaling pathway in some cancer types [47]. Since, the miRNAs are sensitive, specific, and noninvasive, they can be introduced as efficient biomarkers of tumor progression. In present review, for the first time we have summarized all of the miRNAs that have been involved in regulation of WNT signaling pathway in urothelial and prostate cancers to pave the way of introducing a noninvasive diagnostic panel of WNT related miRNAs in these tumors (Table 1, Fig. 1).

Fig. 1
figure 1

Role of miRNAs in regulation of WNT signaling pathway in bladder and prostate cancers. Red and green arrows refer to the regulatory functions in prostate and bladder cancers, respectively

Prostate cancer

The WNT signaling is a critical pathway during PCa progression in which the Wnt signals induce the β-catenin nuclear accumulation resulting in up regulation of WNT target genes [53]. Normally, the destruction complex (Axin/APC/GSK-3β/CK1) maintains cytoplasmic β-catenin levels [54]. It has been reported that there were increased levels of miR-182 expressions in PCa tissues compared with normal margins that promoted cell proliferation and invasion. MiR-182 promoted WNT pathway through the targeting of WNT destruction complex components including APC, Axin, CK1, and GSK-3β [55].

Frizzled7 (FZD7) as a co-receptor of Wnt signaling is associated with tumor cell proliferation, metastasis, and EMT [56]. It is coupled with the canonical WNT/b-catenin pathway, which results in disheveled activation, GSK-3 suppression, and nuclear accumulation of b-catenin. It has been reported that there was an inverse correlation between FZD7 and miR-613 expressions in PCa samples. FZD7 functions as a receptor for the WNT ligands that can be targeted by miR-613 in PCa cells. Therefore, miR-613 up regulation inhibited the PCa cell proliferation and invasion by FZD7/WNT signaling suppression [57]. It has been shown that there was a significant SNHG7 over expression in PCa tissues and cell lines which promoted PCa invasion through miR-324-3p sponging to regulate the WNT2B expression [58]. There was significant reduced levels of miR-26a expression in PCa tissues compared with normal margins. It also inhibited PCa progression and in vivo growth through WNT5a suppression [59].

Astrocyte elevated gene-1 (AEG-1) is a regulator of cell proliferation, angiogenesis, and drug resistance in different cancers [60]. AEG-1 is a scaffold protein that activates various signaling pathways such as MEK/ERK, PI3K/Akt, and WNT [61]. It induces the b-catenin nuclear transport that results in EMT through E-cadherin down regulation and Vimentin over expression [62]. It has been shown that there was significant miR-1297 down regulation in PCa tissues which suppressed PCa cell proliferation and invasion through AEG-1 targeting [63]. DIXDC1 is a positive regulator of the WNT signaling pathway which has an important function in formation of Disheveled, Axin, and b-catenin complex in Wnt pathway [64]. It activates the WNT3A signaling via DVL2. It has been reported that there was DIXDC1 up regulation in PCa cell lines. The miR-1271 also suppressed tumor cell proliferation and invasion through DIXDC1 targeting [65].

N-myc downstream-regulated gene 2 (NDRG2) is involved in cell proliferation and apoptosis by regulation of WNT, MAPK, TGFβ, and AKT signaling pathways [66,67,68]. It inhibits the WNT-mediated transcriptional activation of CCND1. It has been reported that there was miR-454 up regulation in PCa tissues and cell lines. There was also a significant inverse correlation between NDRG2 and miR-454 expressions in PCa tissues. Moreover, miR-454 down regulation inhibited the WNT signaling [69]. DKK3 is an inhibitor of WNT signaling through suppression of LRP5/6 interaction with WNT that induces internalization of LRP5/6. DKK3 and SMAD4 up regulations reduced the tumorigenicity of PCa cells through WNT signaling. There were significant increased levels of miR-183 expressions in PCa tissues in comparison with normal margins which were directly correlated with prostate-specific antigen (PSA) and pT stage, while inversely associated with overall survival. MiR-183 activated the WNT pathway via DKK3 and SMAD4 targeting in PCa cells [70]. NKD1 is another negative regulator of WNT signaling via β-catenin suppression [71]. It has been reported that there were increased levels of miR-744 expressions in castration-resistant prostate cancer (CRPC) compared with androgen-dependent prostate cancer (ADPC) samples which was inversely correlated with CRPC patients survival. MiR-744 promoted cell proliferation and migration through down regulation of WNT signaling inhibitors such as SFRP1, GSK3β, and NDK1 in CRPC cells [72].

Cancer stem cells (CSCs) are a subpopulation of tumor cells with self-renewal ability and chemo radio therapeutic resistance that are one of the main reasons of tumor relapse [73, 74]. Therefore, a combination therapy including chemotherapy, radiotherapy, and surgical resection could be efficient for the CSCs elimination [75]. The sphere-forming CSCs have an extensive proliferation rate and resistance toward the chemotherapy that maintain tumor growth [76]. GSK3β is one of the components of destruction complex that is responsible for β-Catenin phosphorylation and ubiquitination. Since, β-Catenin degradation inactivates WNT pathway, the GSK3β inhibition promotes WNT signaling [77]. SFRP1 is also a negative regulator of WNT pathway [78]. SFRP family contains a homologous domain to the WNT-binding site of Frizzled that prevents the WNT-FZ interaction. It has been reported that there were miR-1301-3p up regulations in PCa tissues that promoted the PCa stem cells through GSK3β and SFRP1 targeting. MiR-1301-3p up regulation also increased the sphere formation [79]. Another study reported that there were significant lower levels of miR-320 expressions in PCa tissues in comparison with normal margins. MiR-320 inhibited the β-catenin and self-renewal ability [80]. The reduced levels of miR-138 expressions have been shown in highly aggressive in comparison with less metastatic cell lines. MiR-138 also impaired cell invasion via E-cadherin up regulation and vimentin down regulation. Moreover, miR-138 inhibits the β-catenin to maintain PCa cell epithelial status [81].

Transcription factor 7 (TCF7) belongs to the TCF/LEF family that is a key effector of WNT pathway [82]. Although, Androgen deprivation therapy (ADT) is an important therapeutic method of PCa via Androgen Receptor (AR) blocking [83], castration-resistant prostate cancer (CRPC) obtains multiple mutations resulting in ADT resistance [84]. WNT signaling is one of the main alternative signaling pathways for the AR signaling in CRPC [85]. There was TCF7 up regulation in advanced PCa tumors that was inversely associated with miR-1. AR inhibited WNT signaling via miR-1-mediated suppression of TCF7 which prohibited the β-catenin–TCF7 complex formation [86]. Ras signaling is also a critical pathway during PCa metastasis [87]. It mediates its functions through induction of various pathways such as MAPK, PI3K/AKT, and WNT signaling [88, 89]. Ras and WNT cooperation is involved in invasive PCa by COX-2 and c-MYC up regulations [90, 91]. It has been shown that the Ras signaling down regulated the miR-34a which is an inhibitor of WNT signaling through TCF7 targeting [92]. PYGO2 is the main component of WNT transcriptional machinery. It has been shown that there was an inverse correlation between the levels of lncRNA625 and miR-432 expressions in the PCa tissues in comparison with normal tissues. lncRNA625 up regulation inhibited the tumor cells growth and induced apoptosis. MiR-432 inactivated the WNT pathway through TRIM29 and PYGO2 targeting. Therefore, lncRNA625 inhibited PCa cells proliferation through activation of WNT pathway by miR-432 targeting [93]. LEF1 is also one of the components of WNT transcriptional machinery that regulates the EMT through interaction with Twist1 and Snail transcription factors [94]. It mediates transcription response to the canonical WNT pathway via β-catenin interaction in which it regulates the expression of WNT target genes such as Cyclin D1 and c-MYC [95]. It has been shown that there was an inverse association between miR-34a and LEF1 expression in PCa cell lines and tissues. MiR-34a regulated the EMT process through LEF1 targeting [96]. SOX is a family of transcription factors involved in various biological processes [97]. SOX30 has a critical role in activation of post-meiotic genes during spermiogenesis. SOX30 inhibits the WNT signaling through CTNNB1 down regulation and preventing the CTNNB1-TCF7L2/TCF4 interaction [98]. It has been reported that there were significant decreased SOX30 levels in PCa cells compared with normal cells. SOX30 up regulation significantly decreased PCa cell proliferation and invasion. There was also an inverse correlation between SOX30 and miR-653-5p in which the SOX30 overexpression or miR-653-5p down regulation inhibited PCa cell proliferation through WNT signaling inactivation [99].

LGR4 is a G protein-coupled receptor (GPCR) that has pivotal roles in tissue development and tumor progression through induction of PI3K/AKT and WNT signaling pathways [100]. It activates the WNT pathway through binding with R-spondins. There is a positive correlation between the IL-6 and LGR4 expressions in tumor cells [101]. It has been reported that the miR-218 suppressed PCa cell proliferation and invasion via LGR4 targeting. MiR-218 also reduced PI3K/Akt phosphorylation and β-catenin accumulation. Moreover, it down regulated the cyclin A1 and MMP-9 via PI3K/AKT and WNT signaling pathways in PCa cells [102]. Calpains belong to the calcium-dependent cysteine proteases involved in apoptosis, cell proliferation, and invasion [103]. CAPN4 regulates the tumor cell proliferation and invasion by WNT signaling activation [104]. It has been shown that there was a high level of CAPN4 expression in PCa cell lines that promotes cell invasion by WNT activation. The miR-520b inhibited the PCa cells invasion via CAPN4 targeting [105]. Zinc is a critical element of cellular proteins involved in cell differentiation and DNA synthesis, whose homeostasis is related with zinc transporters [106]. SLC30/ZnT and SLC39/ZIP are the main zinc transporters in mammalians [107]. SLC39A7 also regulates zinc-mediated tyrosine kinase signaling during tumor progression [108]. It is a Zinc transporter in endoplasmic reticulum/Golgi apparatus that is induced by Epidermal growth factor (EGF), Ca2+, and exogenous Zn2+. It has been reported that there were significant miR-15a-3p down regulations in PCa samples and cell lines. MiR-15a-3p suppressed PCa cell proliferation and invasion through WNT1, b-catenin, and c-MYC targeting. It also up regulated E-cadherin, while down regulated Vimentin via WNT signaling suppression. MiR-15a-3p inhibited the WNT signaling via SLC39A7 targeting [109]. As an intermediate filament, the Keratin 5 (KRT5) is expressed in basal layer stratified epithelial cells [110]. It has been reported that the miR-601 down regulation inhibited the prostate cancer stem cells (PCSCs) proliferation and invasion through KRT5 induction and WNT signaling suppression. The miR-601 down regulation inhibited the NANOG and OCT4 expressions. It also significantly increased the levels of Wnt1 and β-catenin expressions which showed the effect of miR-601/KRT5/WNT on PCSCs [111].

Bladder cancer

WNT5A as a non-canonical WNT is involved in β-catenin phosphorylation and canonical WNT signaling suppression [112]. WNT5A-FZD2 interaction activates STAT3 and promotes EMT process. WNT5 is also involved in GSK3b inhibition which results in b-catenin stabilization [113]. Although, WNT5A promotes non-canonical WNT pathways, it can suppress the canonical WNT pathway via β-catenin degradation [114]. It has been observed that there was a positive association between the levels of miR-374a expression and prognosis in NMIBC patients. Since the over expressed miR-374a patients had longer relapse-free survival (RFS), miR-374a can be introduced as an inhibitor of aggressive tumor through WNT5A targeting and canonical WNT signaling blockade in BCa patients [115]. Gemcitabine is an inhibitor of DNA synthesis which is an efficient neoadjuvant chemotherapy drug to improve the survival of MIBC patients [116]. It has been reported that there was a correlation between miR-129-5p down regulation and gemcitabine resistance in BCa cells. It increased gemcitabine sensitivity in BCa cells through Wnt5a targeting [117]. Higher levels of Wnt7a expressions were observed in 5637 HMI cells compared with 5637 NMI cells. The Wnt7a up regulation was significantly correlated with UBC cell invasiveness and poor prognosis in UBC patients. MiR-370-3p/Wnt7a axis regulated UBC invasion via canonical WNT signaling in BCa [118].

The β-catenin is a pivotal positive regulator of WNT signaling pathway. TRIM29 induces the WNT signaling through β-catenin up regulation, stabilization, and decreasing its phosphorylation level [119, 120]. It has been shown that there were lower levels of miR-621 expressions in BCa tissues compared with normal margins which were negatively associated with overall survival. MiR-621 suppressed the BCa cell proliferation and metastasis through TRIM29 targeting and subsequent WNT signaling inhibition [121]. There were significant miR-1826 down regulations in BC tissues that function as a tumor suppressor through CTNNB1 and VEGFC targeting [122]. It has been shown that there were reduced levels of miR-3619 expressions in BCa cell lines and tissues. MiR-3619 had an important role during BCa progression and metastasis through CDK2 and β-catenin targeting. It also up regulated the p21 that is a key factor in BCa suppression. Moreover, miR-3619 down regulated the c-MYC via reduction in cytoplasmic β-catenin, which suggested the miR-3619 as an inhibitor of WNT pathway in BCa cells. MiR-3619 up regulated the E-cadherin and down regulated the Vimentin and N-cadherin that showed miR-3619 inhibited the EMT process in BCa [123].

GSK-3β is a suppressor of WNT signaling pathway via β-catenin phosphorylation and degradation. It has been reported that there was a significant converse association between the miR-9 and GSK-3β expressions in BCa tissues [124]. AXIN2 is one of the components of destruction complex in WNT pathway. It has been shown that there were increased levels of miR-183 expression in BCa samples that induced tumor cell proliferation. It activated the WNT signaling via AXIN2 suppression [125].

High mobility group box 3 (HMGB3) has an important role in DNA replication and transcription [126]. HMGB3 also inhibits the immune cell differentiation and regulates the hematopoietic stem cells self-renewal and differentiation. There are low levels of HMGB3 expressions in adult tissues, while there are HMGB3 up regulations during self-renewal of hematopoietic stem cells [127]. HMGB3 is a positive regulator of WNT/β-catenin signaling that induces the β-catenin expression and WNT/β-catenin activation [128]. It has been reported that there were decreased levels of miR-532-5p expressions in BCa tissues. MiR-532-5p suppressed the BCa cell proliferation and invasion through HMGB3 targeting and WNT/β-catenin signaling inhibition [129]. EZH2 is one of the components of Polycomb repressive complex 2 that suppress the gene expressions via histone H3 trimethylation at lysine 27 (H3K27me3) [130]. It has been reported that the miR-144 down regulation resulted in WNT signaling activation and BCa cell proliferation by EZH2 suppression. There were also significant lower levels of miR-144 expressions in BCa tissues and cell lines. Furthermore, EZH2 induced H3K27me3 in NKD1 and APC promoter sequences leading to NKD1 and APC down regulations and subsequent WNT signaling activation [131].

Table 1 All of the microRNAs associated with regulation of WNT signaling in prostate and bladder cancers

Conclusion

WNT signaling pathway has a pivotal role during embryogenesis and tumor progression. It has been reported that the miRNAs are one of the main regulators of WNT signaling. In present review, we have summarized all of the miRNAs that have been associated with regulation of WNT signaling pathway during urothelial and prostate tumor progression and metastasis. It was observed that miRNAs mainly regulated the WNT signaling in BCa cells through targeting the WNT ligands and cytoplasmic WNT components such as WNT5A, WNT7A, CTNNB1, GSK3β, and AXIN. While, miRNAs mainly affected the WNT signaling in PCa cells via targeting the cytoplasmic WNT components and WNT related transcription factors such as CTNNB1, GSK3β, AXIN, TCF7, and LEF1. Majority of the reported miRNAs had a tumor suppressor function in bladder and prostate tumor cells by inhibition of the WNT signaling pathway. This review paves the way of introducing a noninvasive diagnostic WNT related miRNAs panel markers in UC and PCa for the first time in the world.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

ADT:

Androgen deprivation therapy

AR:

Androgen receptors

ADPC:

Androgen-dependent prostate cancer

AEG-1:

Astrocyte elevated gene-1

BCa:

Bladder cancer

CSCs:

Cancer stem cells

CRPC:

Castration-resistant prostate cancer

EGF:

Epidermal growth factor

EMT:

Epithelial-mesenchymal transition

ERs:

Estrogen receptors

Fzd7:

Frizzled7

GPCR:

G protein-coupled receptor

GSK-3:

Glycogen synthase kinase-3

H3K27me3:

Histone H3 trimethylation at lysine 27

KRT5:

Keratin 5

LEF:

Lymphoid enhancer-binding factor

mPCa:

Metastatic prostate cancer

miRNAs:

MicroRNAs

MDR:

Multi-drug resistance

MIBC:

Muscle-invasive bladder cancer

MIUC:

Muscle-invasive urothelial carcinoma

NDRG2:

N-myc downstream-regulated gene 2

NMIBC:

Non-muscle invasive bladder cancer

PLDN:

Pelvic lymphadenectomy

PCa:

Prostate cancer

PCSCs:

Prostate cancer stem cells

PSA:

Prostate-specific antigen

RT:

Radiation therapy

RP:

Radical prostatectomy

RFS:

Relapse-free survival

TCF:

T cell-specific factor

TCF7:

Transcription factor 7

UC:

Urothelial cancer

UCB:

Urothelial carcinoma of the bladder

References

  1. Miller KD, Goding Sauer A, Ortiz AP, Fedewa SA, Pinheiro PS, Tortolero-Luna G et al (2018) Cancer statistics for hispanics/latinos, 2018. CA Cancer J Clin 68(6):425–445

    Article  PubMed  Google Scholar 

  2. Radkiewicz C, Edgren G, Johansson ALV, Jahnson S, Häggström C, Akre O et al (2020) Sex differences in Urothelial bladder cancer survival. Clin Genitourin Cancer 18(1):26–34

    Article  PubMed  Google Scholar 

  3. Lin H-J, Sheu PC-Y, Tsai JJ, Wang CC, Chou C-Y (2020) Text mining in a literature review of urothelial cancer using topic model. BMC Cancer 20:1–7

    Article  CAS  Google Scholar 

  4. Pernar CH, Ebot EM, Wilson KM, Mucci LA (2018) The epidemiology of prostate cancer. Cold Spring Harb Perspect Med 8(12):a030361

    Article  PubMed Central  PubMed  Google Scholar 

  5. Häggström C, Stocks T, Nagel G, Manjer J, Bjørge T, Hallmans G et al (2014) Prostate cancer, prostate cancer death, and death from other causes, among men with metabolic aberrations. Epidemiology 25(6):823

    Article  PubMed Central  PubMed  Google Scholar 

  6. Bidarra D, Constâncio V, Barros-Silva D, Ramalho-Carvalho J, Moreira-Barbosa C, Antunes L et al (2019) Circulating microRNAs as biomarkers for prostate cancer detection and metastasis development prediction. Front Oncol 9:900

    Article  PubMed Central  PubMed  Google Scholar 

  7. Schrecengost R, Knudsen KE (eds) (2013) Molecular pathogenesis and progression of prostate cancer. Seminars in oncology. Elsevier

    Google Scholar 

  8. Heidenreich A, Bastian PJ, Bellmunt J, Bolla M, Joniau S, van der Kwast T et al (2014) EAU guidelines on prostate cancer. Part II: Treatment of advanced, relapsing, and castration-resistant prostate cancer. Eur Urol 65(2):467–479

    Article  CAS  PubMed  Google Scholar 

  9. Shelley M, Harrison C, Coles B, Staffurth J, Wilt TJ, Mason MD (2006) Chemotherapy for hormone-refractory prostate cancer. Cochr Database System Rev 2006(4):CD005247

    Google Scholar 

  10. Zhang Q, Cheng H, Wang Y, Tian Y, Xia J, Wang Y et al (2019) Different therapeutic regimens in the treatment of metastatic prostate cancer by performing a Bayesian network meta-analysis. Int J Surg 66:28–36

    Article  PubMed  Google Scholar 

  11. Chen F-z, Zhao X-k (2013) Prostate cancer: current treatment and prevention strategies. Iran Red Crescent Med J 15(4):279

    Article  PubMed Central  PubMed  Google Scholar 

  12. Saginala K, Barsouk A, Aluru JS, Rawla P, Padala SA, Barsouk A (2020) Epidemiology of bladder cancer. Med Sci 8(1):15

    CAS  Google Scholar 

  13. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A et al (2021) Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 71(3):209–249

    Article  PubMed  Google Scholar 

  14. Garg M, Maurya N (2019) WNT/β-catenin signaling in urothelial carcinoma of bladder. World J Nephrol 8(5):83

    Article  PubMed Central  PubMed  Google Scholar 

  15. Burger M, Catto JW, Dalbagni G, Grossman HB, Herr H, Karakiewicz P et al (2013) Epidemiology and risk factors of urothelial bladder cancer. Eur Urol 63(2):234–241

    Article  PubMed  Google Scholar 

  16. Mojarrad M, Moghbeli M (2020) Genetic and molecular biology of bladder cancer among Iranian patients. Mol Genet Genomic Med 8(6):e1233

    Article  PubMed Central  PubMed  Google Scholar 

  17. Ishida K, Hsieh MH (2018) Understanding urogenital schistosomiasis-related bladder cancer: an update. Front Med (Lausanne) 5:223

    Article  Google Scholar 

  18. Miyamoto H, Yang Z, Chen Y-T, Ishiguro H, Uemura H, Kubota Y et al (2007) Promotion of bladder cancer development and progression by androgen receptor signals. J Natl Cancer Inst 99(7):558–568

    Article  CAS  PubMed  Google Scholar 

  19. Li P, Chen J, Miyamoto H (2017) Androgen receptor signaling in bladder cancer. Cancers 9(2):20

    Article  PubMed Central  CAS  Google Scholar 

  20. Tripathi A, Gupta S (2020) Androgen receptor in bladder cancer: a promising therapeutic target. Asian J Urol 7(3):284–290

    Article  PubMed Central  PubMed  Google Scholar 

  21. Goto T, Miyamoto H (2021) The role of estrogen receptors in urothelial cancer. Front Endocrinol 12:246

    Article  Google Scholar 

  22. Di Pierro GB, Gulia C, Cristini C, Fraietta G, Marini L, Grande P et al (2012) Bladder cancer: a simple model becomes complex. Curr Genomics 13(5):395

    Article  PubMed Central  PubMed  Google Scholar 

  23. Schulz WA (2006) Understanding urothelial carcinoma through cancer pathways. Int J Cancer 119(7):1513–1518

    Article  CAS  PubMed  Google Scholar 

  24. Moschini M, D’andrea D, Korn S, Irmak Y, Soria F, Compérat E et al (2017) Characteristics and clinical significance of histological variants of bladder cancer. Nat Rev Urol 14(11):651–668

    Article  PubMed  Google Scholar 

  25. Chalasani V, Chin JL, Izawa JI (2009) Histologic variants of urothelial bladder cancer and nonurothelial histology in bladder cancer. Can Urol Assoc J 3(6 Suppl 4):S193

    PubMed Central  PubMed  Google Scholar 

  26. Sylvester RJ, Rodríguez O, Hernández V, Turturica D, Bauerová L, Bruins HM et al (2021) European Association of Urology (EAU) prognostic factor risk groups for non–muscle-invasive bladder cancer (NMIBC) incorporating the WHO 2004/2016 and WHO 1973 classification systems for grade: an update from the EAU NMIBC Guidelines Panel. Eur Urol 79(4):480–488

    Article  PubMed  Google Scholar 

  27. Ramos P, Pereira P, Dinis P, Pacheco-Figueiredo L (2021) Bladder cancer variant histologies: epidemiology, diagnosis, treatment and prognosis. modern approach to diagnosis and treatment of bladder cancer. IntechOpen

    Google Scholar 

  28. Marcos-Gragera R, Mallone S, Kiemeney LA, Vilardell L, Malats N, Allory Y et al (2015) Urinary tract cancer survival in Europe 1999–2007: results of the population-based study EUROCARE-5. Eur J Cancer 51(15):2217–2230

    Article  PubMed  Google Scholar 

  29. Pasin E, Josephson DY, Mitra AP, Cote RJ, Stein JP (2008) Superficial bladder cancer: an update on etiology, molecular development, classification, and natural history. Rev Urol 10(1):31–43

    PubMed Central  PubMed  Google Scholar 

  30. Liebert M, Seigne J (1996) Characteristics of invasive bladder cancers: histological and molecular markers. Semin Urol Oncol 14(2):62–72

    CAS  PubMed  Google Scholar 

  31. Stenzl A, Cowan NC, De Santis M, Kuczyk MA, Merseburger AS, Ribal MJ et al (2011) Treatment of muscle-invasive and metastatic bladder cancer: update of the EAU guidelines. Eur Urol 59(6):1009–1018

    Article  CAS  PubMed  Google Scholar 

  32. Dimov I, Visnjic M, Stefanovic V (2010) Urothelial cancer stem cells. TheScientificWorldJOURNAL 10:1400–1415

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  33. Sheikh A, Niazi AK, Ahmed MZ, Iqbal B, Anwer SMS, Khan HH (2014) The role of Wnt signaling pathway in carcinogenesis and implications for anticancer therapeutics. Hereditary Cancer Clin Pract 12(1):13

    Article  CAS  Google Scholar 

  34. Abbaszadegan MR, Riahi A, Forghanifard MM, Moghbeli M (2018) WNT and NOTCH signaling pathways as activators for epidermal growth factor receptor in esophageal squamous cell carcinoma. Cell Mol Biol Lett 23:42

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  35. Moghbeli M, Abbaszadegan MR, Golmakani E, Forghanifard MM (2016) Correlation of Wnt and NOTCH pathways in esophageal squamous cell carcinoma. J Cell Commun Signal 10(2):129–135

    Article  PubMed Central  PubMed  Google Scholar 

  36. Moghbeli M, Sadrizadeh A, Forghanifard MM, Mozaffari HM, Golmakani E, Abbaszadegan MR (2016) Role of Msi1 and PYGO2 in esophageal squamous cell carcinoma depth of invasion. J Cell Commun Signal 10(1):49–53

    Article  PubMed  Google Scholar 

  37. Zhan T, Rindtorff N, Boutros M (2017) Wnt signaling in cancer. Oncogene 36(11):1461–1473

    Article  CAS  PubMed  Google Scholar 

  38. Grainger S, Willert K (2018) Mechanisms of Wnt signaling and control. Wiley Interdiscip Rev Syst Biol Med 10(5):e1422

    Article  Google Scholar 

  39. Lamouille S, Xu J, Derynck R (2014) Molecular mechanisms of epithelial–mesenchymal transition. Nat Rev Mol Cell Biol 15(3):178–196

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  40. Yang X, Li L, Huang Q, Xu W, Cai X, Zhang J et al (2015) Wnt signaling through Snail1 and Zeb1 regulates bone metastasis in lung cancer. Am J Cancer Res 5(2):748

    PubMed Central  PubMed  Google Scholar 

  41. Majid S, Saini S, Dahiya R (2012) Wnt signaling pathways in urological cancers: past decades and still growing. Mol Cancer 11(1):7

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  42. Murillo-Garzón V, Kypta R (2017) WNT signalling in prostate cancer. Nat Rev Urol 14(11):683

    Article  CAS  PubMed  Google Scholar 

  43. Schatton T, Frank NY, Frank MH (2009) Identification and targeting of cancer stem cells. BioEssays 31(10):1038–1049

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  44. Bao B, Ahmad A, Azmi AS, Ali S, Sarkar FH (2013) Overview of cancer stem cells (CSCs) and mechanisms of their regulation: implications for cancer therapy. Curr Protoc Pharmacol 61(1):14251–142514

    Article  Google Scholar 

  45. Phi LTH, Sari IN, Yang Y-G, Lee S-H, Jun N, Kim KS et al (2018) Cancer stem cells (CSCs) in drug resistance and their therapeutic implications in cancer treatment. Stem Cells Int 2018:5416923

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  46. Yang L, Shi P, Zhao G, Xu J, Peng W, Zhang J et al (2020) Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther 5(1):1–35

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  47. Onyido EK, Sweeney E, Nateri AS (2016) Wnt-signalling pathways and microRNAs network in carcinogenesis: experimental and bioinformatics approaches. Mol Cancer 15(1):1–17

    Article  CAS  Google Scholar 

  48. Kalluri R, Weinberg RA (2009) The basics of epithelial-mesenchymal transition. J Clin Investig 119(6):1420–1428

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  49. Forghanifard MM, Rad A, Farshchian M, Khaleghizadeh M, Gholamin M, Moghbeli M et al (2017) TWIST1 upregulates the MAGEA4 oncogene. Mol Carcinog 56(3):877–885

    Article  CAS  PubMed  Google Scholar 

  50. Zhang J, Tian XJ, Xing J (2016) Signal transduction pathways of EMT induced by TGF-beta, SHH, and WNT and their crosstalks. J Clin Med 5(4):41

    Article  PubMed Central  CAS  Google Scholar 

  51. Su WL, Kleinhanz RR, Schadt EE (2011) Characterizing the role of miRNAs within gene regulatory networks using integrative genomics techniques. Mol Syst Biol 7(1):490

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  52. Peng Y, Zhang X, Feng X, Fan X, Jin Z (2017) The crosstalk between microRNAs and the Wnt/β-catenin signaling pathway in cancer. Oncotarget 8(8):14089

    Article  PubMed  Google Scholar 

  53. Cristobal I, Rojo F, Madoz-Gurpide J, Garcia-Foncillas J (2016) Cross talk between wnt/beta-catenin and CIP2A/Plk1 signaling in prostate cancer: promising therapeutic implications. Mol Cell Biol 36(12):1734–1739

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  54. Ikeda S, Kishida S, Yamamoto H, Murai H, Koyama S, Kikuchi A (1998) Axin, a negative regulator of the Wnt signaling pathway, forms a complex with GSK-3beta and beta-catenin and promotes GSK-3beta-dependent phosphorylation of beta-catenin. EMBO J 17(5):1371–1384

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  55. Wang D, Lu G, Shao Y, Xu D (2018) MiR-182 promotes prostate cancer progression through activating Wnt/beta-catenin signal pathway. Biomed Pharmacother Biomed Pharmacother 99:334–339

    Article  CAS  PubMed  Google Scholar 

  56. Deng B, Zhang S, Miao Y, Zhang Y, Wen F, Guo K (2015) Down-regulation of Frizzled-7 expression inhibits migration, invasion, and epithelial-mesenchymal transition of cervical cancer cell lines. Med Oncol 32(4):102

    Article  CAS  PubMed  Google Scholar 

  57. Ren W, Li C, Duan W, Du S, Yang F, Zhou J et al (2016) MicroRNA-613 represses prostate cancer cell proliferation and invasion through targeting Frizzled7. Biochem Biophys Res Commun 469(3):633–638

    Article  CAS  PubMed  Google Scholar 

  58. Han Y, Hu H, Zhou J (2019) Knockdown of LncRNA SNHG7 inhibited epithelial-mesenchymal transition in prostate cancer though miR-324-3p/WNT2B axis in vitro. Pathol Res Pract 215(10):152537

    Article  CAS  PubMed  Google Scholar 

  59. Zhao S, Ye X, Xiao L, Lian X, Feng Y, Li F et al (2014) MiR-26a inhibits prostate cancer progression by repression of Wnt5a. Tumour Biol J Int Soc Oncodev Biol Med 35(10):9725–9733

    Article  CAS  Google Scholar 

  60. Hu G, Wei Y, Kang Y (2009) The multifaceted role of MTDH/AEG-1 in cancer progression. Clini Cancer Res Off J Am Assoc Cancer Res 15(18):5615–5620

    Article  CAS  Google Scholar 

  61. Robertson CL, Srivastava J, Rajasekaran D, Gredler R, Akiel MA, Jariwala N et al (2015) The role of AEG-1 in the development of liver cancer. Hepatic Oncol 2(3):303–312

    Article  Google Scholar 

  62. He W, He S, Wang Z, Shen H, Fang W, Zhang Y et al (2015) Astrocyte elevated gene-1(AEG-1) induces epithelial-mesenchymal transition in lung cancer through activating Wnt/beta-catenin signaling. BMC Cancer 15:107

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  63. Liang X, Li H, Fu D, Chong T, Wang Z, Li Z (2016) MicroRNA-1297 inhibits prostate cancer cell proliferation and invasion by targeting the AEG-1/Wnt signaling pathway. Biochem Biophys Res Commun 480(2):208–214

    Article  CAS  PubMed  Google Scholar 

  64. Shiomi K, Uchida H, Keino-Masu K, Masu M (2003) Ccd1, a novel protein with a DIX domain, is a positive regulator in the Wnt signaling during zebrafish neural patterning. Curr Biol CB 13(1):73–77

    Article  CAS  PubMed  Google Scholar 

  65. Zhong J, Liu Y, Xu Q, Yu J, Zhang M (2017) Inhibition of DIXDC1 by microRNA-1271 suppresses the proliferation and invasion of prostate cancer cells. Biochem Biophys Res Commun 484(4):794–800

    Article  CAS  PubMed  Google Scholar 

  66. Kim YJ, Yoon SY, Kim JT, Song EY, Lee HG, Son HJ et al (2009) NDRG2 expression decreases with tumor stages and regulates TCF/beta-catenin signaling in human colon carcinoma. Carcinogenesis 30(4):598–605

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  67. Lee DC, Kang YK, Kim WH, Jang YJ, Kim DJ, Park IY et al (2008) Functional and clinical evidence for NDRG2 as a candidate suppressor of liver cancer metastasis. Can Res 68(11):4210–4220

    Article  CAS  Google Scholar 

  68. Park Y, Shon SK, Kim A, Kim KI, Yang Y, Cho DH et al (2007) SOCS1 induced by NDRG2 expression negatively regulates STAT3 activation in breast cancer cells. Biochem Biophys Res Commun 363(2):361–367

    Article  CAS  PubMed  Google Scholar 

  69. Fu Q, Gao Y, Yang F, Mao T, Sun Z, Wang H et al (2018) Suppression of microRNA-454 impedes the proliferation and invasion of prostate cancer cells by promoting N-myc downstream-regulated gene 2 and inhibiting WNT/beta-catenin signaling. Biomed Pharmacother Biomed Pharmacother 97:120–127

    Article  CAS  PubMed  Google Scholar 

  70. Ueno K, Hirata H, Shahryari V, Deng G, Tanaka Y, Tabatabai ZL et al (2013) microRNA-183 is an oncogene targeting Dkk-3 and SMAD4 in prostate cancer. Br J Cancer 108(8):1659–1667

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  71. Angonin D, Van Raay TJ (2013) Nkd1 functions as a passive antagonist of Wnt signaling. PLoS ONE 8(8):e74666

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  72. Guan H, Liu C, Fang F, Huang Y, Tao T, Ling Z et al (2017) MicroRNA-744 promotes prostate cancer progression through aberrantly activating Wnt/beta-catenin signaling. Oncotarget 8(9):14693–14707

    Article  PubMed Central  PubMed  Google Scholar 

  73. Moghbeli M, Moghbeli F, Forghanifard MM, Abbaszadegan MR (2014) Cancer stem cell detection and isolation. Med Oncol 31(9):69

    Article  CAS  PubMed  Google Scholar 

  74. Moghbeli M, Mosannen Mozaffari H, Memar B, Forghanifard MM, Gholamin M, Abbaszadegan MR (2019) Role of MAML1 in targeted therapy against the esophageal cancer stem cells. J Transl Med 17(1):126

    Article  PubMed Central  PubMed  Google Scholar 

  75. Chen X, Liao R, Li D, Sun J (2017) Induced cancer stem cells generated by radiochemotherapy and their therapeutic implications. Oncotarget 8(10):17301–17312

    Article  PubMed  Google Scholar 

  76. Cao L, Zhou Y, Zhai B, Liao J, Xu W, Zhang R et al (2011) Sphere-forming cell subpopulations with cancer stem cell properties in human hepatoma cell lines. BMC Gastroenterol 11:71

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  77. Fodde R, Smits R, Clevers H (2001) APC, signal transduction and genetic instability in colorectal cancer. Nat Rev Cancer 1(1):55–67

    Article  CAS  PubMed  Google Scholar 

  78. Caldwell GM, Jones C, Gensberg K, Jan S, Hardy RG, Byrd P et al (2004) The Wnt antagonist sFRP1 in colorectal tumorigenesis. Can Res 64(3):883–888

    Article  CAS  Google Scholar 

  79. Song XL, Huang B, Zhou BW, Wang C, Liao ZW, Yu Y et al (2018) miR-1301–3p promotes prostate cancer stem cell expansion by targeting SFRP1 and GSK3beta. Biomed Pharmacother Biomed Pharmacother 99:369–374

    Article  CAS  PubMed  Google Scholar 

  80. Hsieh IS, Chang KC, Tsai YT, Ke JY, Lu PJ, Lee KH et al (2013) MicroRNA-320 suppresses the stem cell-like characteristics of prostate cancer cells by downregulating the Wnt/beta-catenin signaling pathway. Carcinogenesis 34(3):530–538

    Article  CAS  PubMed  Google Scholar 

  81. Yu Z, Wang Z, Li F, Yang J, Tang L (2018) miR138 modulates prostate cancer cell invasion and migration via Wnt/betacatenin pathway. Mol Med Rep 17(2):3140–3145

    CAS  PubMed  Google Scholar 

  82. Cadigan KM, Waterman ML (2012) TCF/LEFs and Wnt signaling in the nucleus. Cold Spring Harb Persp Biol 4(11):a007906

    Google Scholar 

  83. Yuan X, Cai C, Chen S, Chen S, Yu Z, Balk SP (2014) Androgen receptor functions in castration-resistant prostate cancer and mechanisms of resistance to new agents targeting the androgen axis. Oncogene 33(22):2815–2825

    Article  CAS  PubMed  Google Scholar 

  84. Karantanos T, Corn PG, Thompson TC (2013) Prostate cancer progression after androgen deprivation therapy: mechanisms of castrate resistance and novel therapeutic approaches. Oncogene 32(49):5501–5511

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  85. Wang G, Wang J, Sadar MD (2008) Crosstalk between the androgen receptor and beta-catenin in castrate-resistant prostate cancer. Can Res 68(23):9918–9927

    Article  CAS  Google Scholar 

  86. Siu MK, Chen WY, Tsai HY, Chen HY, Yin JJ, Chen CL et al (2017) TCF7 is suppressed by the androgen receptor via microRNA-1-mediated downregulation and is involved in the development of resistance to androgen deprivation in prostate cancer. Prostate Cancer Prostatic Dis 20(2):172–178

    Article  CAS  PubMed  Google Scholar 

  87. Carter BS, Epstein JI, Isaacs WB (1990) ras gene mutations in human prostate cancer. Can Res 50(21):6830–6832

    CAS  Google Scholar 

  88. Anastas JN, Moon RT (2013) WNT signalling pathways as therapeutic targets in cancer. Nat Rev Cancer 13(1):11–26

    Article  CAS  PubMed  Google Scholar 

  89. Jiang H, Grenley MO, Bravo MJ, Blumhagen RZ, Edgar BA (2011) EGFR/Ras/MAPK signaling mediates adult midgut epithelial homeostasis and regeneration in Drosophila. Cell Stem Cell 8(1):84–95

    Article  CAS  PubMed  Google Scholar 

  90. Araki Y, Okamura S, Hussain SP, Nagashima M, He P, Shiseki M et al (2003) Regulation of cyclooxygenase-2 expression by the Wnt and ras pathways. Can Res 63(3):728–734

    CAS  Google Scholar 

  91. Kerkhoff E, Houben R, Loffler S, Troppmair J, Lee JE, Rapp UR (1998) Regulation of c-myc expression by Ras/Raf signalling. Oncogene 16(2):211–216

    Article  CAS  PubMed  Google Scholar 

  92. Chen WY, Liu SY, Chang YS, Yin JJ, Yeh HL, Mouhieddine TH et al (2015) MicroRNA-34a regulates WNT/TCF7 signaling and inhibits bone metastasis in Ras-activated prostate cancer. Oncotarget 6(1):441–457

    Article  PubMed  Google Scholar 

  93. Li JB, Liu F, Zhang BP, Bai WK, Cheng W, Zhang YH et al (2017) LncRNA625 modulates prostate cancer cells proliferation and apoptosis through regulating the Wnt/beta-catenin pathway by targeting miR-432. Eur Rev Med Pharmacol Sci 21(11):2586–2595

    PubMed  Google Scholar 

  94. Ezponda T, Popovic R, Shah MY, Martinez-Garcia E, Zheng Y, Min DJ et al (2013) The histone methyltransferase MMSET/WHSC1 activates TWIST1 to promote an epithelial-mesenchymal transition and invasive properties of prostate cancer. Oncogene 32(23):2882–2890

    Article  CAS  PubMed  Google Scholar 

  95. Shtutman M, Zhurinsky J, Simcha I, Albanese C, D’Amico M, Pestell R et al (1999) The cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway. Proc Natl Acad Sci USA 96(10):5522–5527

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  96. Liang J, Li Y, Daniels G, Sfanos K, De Marzo A, Wei J et al (2015) LEF1 Targeting EMT in Prostate Cancer Invasion Is Regulated by miR-34a. Mol Cancer Res MCR 13(4):681–688

    Article  CAS  PubMed  Google Scholar 

  97. She ZY, Yang WX (2015) SOX family transcription factors involved in diverse cellular events during development. Eur J Cell Biol 94(12):547–563

    Article  CAS  PubMed  Google Scholar 

  98. Han F, Liu WB, Shi XY, Yang JT, Zhang X, Li ZM et al (2018) SOX30 Inhibits Tumor Metastasis through Attenuating Wnt-Signaling via Transcriptional and Posttranslational Regulation of beta-Catenin in Lung Cancer. EBioMedicine 31:253–266

    Article  PubMed Central  PubMed  Google Scholar 

  99. Fu Q, Sun Z, Yang F, Mao T, Gao Y, Wang H (2019) SOX30, a target gene of miR-653-5p, represses the proliferation and invasion of prostate cancer cells through inhibition of Wnt/beta-catenin signaling. Cell Mol Biol Lett 24:71

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  100. Liang F, Yue J, Wang J, Zhang L, Fan R, Zhang H et al (2015) GPCR48/LGR4 promotes tumorigenesis of prostate cancer via PI3K/Akt signaling pathway. Med Oncol 32(3):49

    Article  CAS  PubMed  Google Scholar 

  101. Liu J, Wei W, Guo CA, Han N, Pan JF, Fei T et al (2013) Stat3 upregulates leucine-rich repeat-containing g protein-coupled receptor 4 expression in osteosarcoma cells. Biomed Res Int 2013:310691

    Article  PubMed Central  PubMed  Google Scholar 

  102. Li F, Gu C, Tian F, Jia Z, Meng Z, Ding Y et al (2016) MiR-218 impedes IL-6-induced prostate cancer cell proliferation and invasion via suppression of LGR4 expression. Oncol Rep 35(5):2859–2865

    Article  CAS  PubMed  Google Scholar 

  103. Goll DE, Thompson VF, Li H, Wei W, Cong J (2003) The calpain system. Physiol Rev 83(3):731–801

    Article  CAS  PubMed  Google Scholar 

  104. Wang E, Wang D, Li B, Ma H, Wang C, Guan L et al (2017) Capn4 promotes epithelial-mesenchymal transition in human melanoma cells through activation of the Wnt/beta-catenin pathway. Oncol Rep 37(1):379–387

    Article  PubMed  Google Scholar 

  105. Ren W, Wang D, Li C, Shu T, Zhang W, Fu X (2018) Capn4 expression is modulated by microRNA-520b and exerts an oncogenic role in prostate cancer cells by prom. Biomed Pharmacother Biomed Pharmacother 108:467–475

    Article  CAS  PubMed  Google Scholar 

  106. Franklin RB, Costello LC (2009) The important role of the apoptotic effects of zinc in the development of cancers. J Cell Biochem 106(5):750–757

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  107. Cousins RJ, Liuzzi JP, Lichten LA (2006) Mammalian zinc transport, trafficking, and signals. J Biol Chem 281(34):24085–24089

    Article  CAS  PubMed  Google Scholar 

  108. Hogstrand C, Kille P, Nicholson RI, Taylor KM (2009) Zinc transporters and cancer: a potential role for ZIP7 as a hub for tyrosine kinase activation. Trends Mol Med 15(3):101–111

    Article  CAS  PubMed  Google Scholar 

  109. Cui Y, Yang Y, Ren L, Yang J, Wang B, Xing T et al (2019) miR-15a-3p suppresses prostate cancer cell proliferation and invasion by targeting SLC39A7 Via downregulating wnt/beta-catenin signaling pathway. Cancer Biother Radiopharm 34(7):472–479

    Article  CAS  PubMed  Google Scholar 

  110. Srivastava SS, Alam H, Patil SJ, Shrinivasan R, Raikundalia S, Chaudhari PR et al (2018) Keratin 5/14mediated cell differentiation and transformation are regulated by TAp63 and Notch1 in oral squamous cell carcinomaderived cells. Oncol Rep 39(5):2393–2401

    CAS  PubMed  Google Scholar 

  111. Du H, Wang X, Dong R, Hu D, Xiong Y (2019) miR-601 inhibits proliferation, migration and invasion of prostate cancer stem cells by targeting KRT5 to inactivate the Wnt signaling pathway. Int J Clin Exp Pathol 12(12):4361–4379

    PubMed Central  PubMed  Google Scholar 

  112. Li J, Ying J, Fan Y, Wu L, Ying Y, Chan AT et al (2010) WNT5A antagonizes WNT/beta-catenin signaling and is frequently silenced by promoter CpG methylation in esophageal squamous cell carcinoma. Cancer Biol Ther 10(6):617–624

    Article  CAS  PubMed  Google Scholar 

  113. Torii K, Nishizawa K, Kawasaki A, Yamashita Y, Katada M, Ito M et al (2008) Anti-apoptotic action of Wnt5a in dermal fibroblasts is mediated by the PKA signaling pathways. Cell Signal 20(7):1256–1266

    Article  CAS  PubMed  Google Scholar 

  114. Topol L, Jiang X, Choi H, Garrett-Beal L, Carolan PJ, Yang Y (2003) Wnt-5a inhibits the canonical Wnt pathway by promoting GSK-3-independent beta-catenin degradation. J Cell Biol 162(5):899–908

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  115. Chen X, Jia C, Jia C, Jin X, Gu X (2018) MicroRNA-374a inhibits aggressive tumor biological behavior in bladder carcinoma by suppressing wnt/beta-catenin signaling. Cell Physiol Biochem Int J Exp Cell Physiol Biochem Pharmacol 48(2):815–826

    Article  CAS  Google Scholar 

  116. Anghel RM, Gales LN, Trifanescu OG (2016) Outcome of urinary bladder cancer after combined therapies. J Med Life 9(2):153–159

    CAS  PubMed Central  PubMed  Google Scholar 

  117. Cao J, Wang Q, Wu G, Li S, Wang Q (2018) miR-129-5p inhibits gemcitabine resistance and promotes cell apoptosis of bladder cancer cells by targeting Wnt5a. Int Urol Nephrol 50(10):1811–1819

    Article  CAS  PubMed  Google Scholar 

  118. Huang X, Zhu H, Gao Z, Li J, Zhuang J, Dong Y et al (2018) Wnt7a activates canonical Wnt signaling, promotes bladder cancer cell invasion, and is suppressed by miR-370-3p. J Biol Chem 293(18):6693–6706

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  119. Sun J, Zhang T, Cheng M, Hong L, Zhang C, Xie M et al (2019) TRIM29 facilitates the epithelial-to-mesenchymal transition and the progression of colorectal cancer via the activation of the Wnt/beta-catenin signaling pathway. J Exp Clin Cancer Res CR 38(1):104

    Article  CAS  PubMed  Google Scholar 

  120. Wang L, Heidt DG, Lee CJ, Yang H, Logsdon CD, Zhang L et al (2009) Oncogenic function of ATDC in pancreatic cancer through Wnt pathway activation and beta-catenin stabilization. Cancer Cell 15(3):207–219

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  121. Tian H, Wang X, Lu J, Tian W, Chen P (2019) MicroRNA-621 inhibits cell proliferation and metastasis in bladder cancer by suppressing Wnt/beta-catenin signaling. Chem Biol Interact 308:244–251

    Article  CAS  PubMed  Google Scholar 

  122. Hirata H, Hinoda Y, Ueno K, Shahryari V, Tabatabai ZL, Dahiya R (2012) MicroRNA-1826 targets VEGFC, beta-catenin (CTNNB1) and MEK1 (MAP2K1) in human bladder cancer. Carcinogenesis 33(1):41–48

    Article  CAS  PubMed  Google Scholar 

  123. Zhang Q, Miao S, Han X, Li C, Zhang M, Cui K et al (2018) MicroRNA-3619-5p suppresses bladder carcinoma progression by directly targeting beta-catenin and CDK2 and activating p21. Cell Death Dis 9(10):960

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  124. Dong ZC, Zhang D, Wang SB, Lin ZQ (2018) Target inhibition on GSK-3beta by miR-9 to modulate proliferation and apoptosis of bladder cancer cells. Eur Rev Med Pharmacol Sci 22(10):3018–3026

    PubMed  Google Scholar 

  125. Chen D, Li SG, Chen JY, Xiao M (2018) MiR-183 maintains canonical Wnt signaling activity and regulates growth and apoptosis in bladder cancer via targeting AXIN2. Eur Rev Med Pharmacol Sci 22(15):4828–4836

    CAS  PubMed  Google Scholar 

  126. Stros M (2010) HMGB proteins: interactions with DNA and chromatin. Biochem Biophys Acta 1799(1–2):101–113

    CAS  PubMed  Google Scholar 

  127. Nemeth MJ, Kirby MR, Bodine DM (2006) Hmgb3 regulates the balance between hematopoietic stem cell self-renewal and differentiation. Proc Natl Acad Sci USA 103(37):13783–13788

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  128. Zhang Z, Chang Y, Zhang J, Lu Y, Zheng L, Hu Y et al (2017) HMGB3 promotes growth and migration in colorectal cancer by regulating WNT/beta-catenin pathway. PLoS ONE 12(7):e0179741

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  129. Xie X, Pan J, Han X, Chen W (2019) Downregulation of microRNA-532-5p promotes the proliferation and invasion of bladder cancer cells through promotion of HMGB3/Wnt/beta-catenin signaling. Chem Biol Interact 300:73–81

    Article  CAS  PubMed  Google Scholar 

  130. Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage H, Tempst P et al (2002) Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science 298(5595):1039–1043

    Article  CAS  PubMed  Google Scholar 

  131. Guo Y, Ying L, Tian Y, Yang P, Zhu Y, Wang Z et al (2013) miR-144 downregulation increases bladder cancer cell proliferation by targeting EZH2 and regulating Wnt signaling. FEBS J 280(18):4531–4538

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

Not applicable.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

MM, NT, and MM were involved in search strategy and drafting. MM supervised the project and revised and edited the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Meysam Moghbeli.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Montazer, M., Taghehchian, N., Mojarrad, M. et al. Role of microRNAs in regulation of WNT signaling pathway in urothelial and prostate cancers. Egypt J Med Hum Genet 23, 102 (2022). https://doi.org/10.1186/s43042-022-00315-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43042-022-00315-8

Keywords