Next Article in Journal
Metformin Treatment Modulates Long Non-Coding RNA Isoforms Expression in Human Cells
Previous Article in Journal
Emerging Role of Enhancer RNAs as Potential Diagnostic and Prognostic Biomarkers in Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Roles of microRNA miR-185 in Digestive Tract Cancers

by
Esmaeel Babaeenezhad
1,2,3,
Fakhraddin Naghibalhossaini
4,
Masoumeh Rajabibazl
5,
Zohreh Jangravi
6,
Forouzan Hadipour Moradi
7,
Mohammad Davood Fattahi
3,
Jörg D. Hoheisel
8,
Mostafa Moradi Sarabi
2,9,*,† and
Soroosh Shahryarhesami
8,*,†
1
Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran
2
Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad 6813816314, Iran
3
Department of Clinical Biochemistry, School of Medicine, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran 1983969411, Iran
4
Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
5
Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1983969411, Iran
6
Department of Biochemistry, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran
7
Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad 6813816314, Iran
8
Functional Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
9
Department of Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad 6813816314, Iran
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Non-Coding RNA 2022, 8(5), 67; https://doi.org/10.3390/ncrna8050067
Submission received: 23 August 2022 / Revised: 30 September 2022 / Accepted: 1 October 2022 / Published: 8 October 2022

Abstract

:
Digestive tract cancers represent a serious public health issue. In recent years, evidence has accumulated that microRNA miR-185 is implicated in the pathogenesis of this group of highly malignant tumors. Its expression variations correlate with clinical features, such as tumor size, lymph node metastasis, tumor node metastatic stage, survival, recurrence and response to adjuvant therapy, and have diagnostic and prognostic potential. In this review, we compile, evaluate and discuss the current knowledge about the roles of miR-185 in digestive tract cancers. Interestingly, miR-185 is apparently involved in regulating both tumor suppressive and oncogenic processes. We look at downstream effects as well as upstream regulation. In addition, we discuss the utility of miR-185 for diagnosis and its potential concerning novel therapeutic approaches.

1. Introduction

Digestive tract cancers are considered a major global health problem. They account for about one-third of global cancer cases and cancer-related mortality [1,2]. Overall, they are responsible for about 4.1 million new cases worldwide and lead to about 3 million deaths each year. Despite advances in treatment strategies, many patients still face poor outcomes; the five-year survival rate of gastric cancer (GC) patients, for example, is less than 10%; the mortality of colorectal cancer (CRC) ranks second among malignant tumors [3] but will be replaced by pancreatic cancer at around 2030 [4]. Digestive tract cancer carcinogenesis is associated with genetic and epigenetic changes [5]. Chromosomal instability—originating from chromosome segregation dysfunction, defective DNA damage response, or induced by Helicobacter pylori—as well as microsatellite instability resulting from mismatch repair deficiency are among the most common genetic alterations observed. With respect to epigenetic alterations, aberrant DNA methylation, histone modifications, and altered microRNA (miR) expression are the most important factors involved in pathogenesis [6,7].
Over the years, we have studied the miR-content in cancer tissues and blood samples for both understanding tumor biology and determining the diagnostic potential [8,9,10,11]. miRs are non-protein-encoding RNA molecules that play a critical role in the post-transcriptional regulation of gene expression. The biogenesis of miRs is a multistep process that takes place in the cells’ nucleus and cytoplasm. They are transcribed from intergenic or intronic regions by RNA polymerase II as a primary miRNA and then processed by RNase III enzyme Drosha into stem and loop structures called pre-miRNAs. They are transferred to the cytoplasm and converted to mature double-stranded molecules by the RNase III enzyme Dicer [12,13]. Together with Argonaute proteins, miRs participate in the formation of an RNA-induced silencing complex (RISC), which leads to the degradation or translational suppression of specific target mRNAs. Alteration of miR expression contributes to the development of many diseases. Not surprisingly, miRs are involved in regulating fundamental functions related to cancer, such as cellular proliferation and apoptosis, cell cycle and metabolism, and differentiation [14]. Depending on the physiological environment and pathological condition, miRs may act as oncogenes that induce tumorigenesis or tumor suppressors that restrain tumor development. They also allow a prognosis for different cancer types and have a significant diagnostic and therapeutic potential [15].
MicroRNA miR-185 has been found frequently to vary in abundance in samples from cancer patients compared to samples from healthy donors. The gene is located at chromosome 22q11.21. The miR precursor consists of 82 nucleotides and is the source of the two mature molecules miR-185-5p and miR-185-3p; according to available data, miR-185-5p is the predominantly produced molecule (Figure 1).
Dysregulation of miR-185 has been found in various pathological states, and there is much evidence for the misregulation of miR-185 in human cancers. The majority of experimental data indicates that miR-185 exhibits tumor-suppressive activities by affecting many critical biological processes such as cell cycle, epithelial-to-mesenchymal transition (EMT), apoptosis, autophagy, invasion and metastasis. However, fewer but nevertheless several studies have reported that miR-185 acts as an oncogene.
Because of its apparently central role in tumor-related, miR-based regulation and the lack of a compilation and review of the very many different activities related to miR-185 regulation, we set out to summarize the current knowledge about the roles of miR-185 in digestive tract cancers and the molecular mechanisms that are affected. We look at downstream genes regulated by miR-185 and upstream molecules that are involved in controlling miR-185 expression. In addition, we discuss its utility for diagnosis and the potential for therapy. For simplicity, we ordered the review according to the location of the respective tissue in the body, starting with oral cancer and finishing with colorectal tumors. For an overview, a graphical synopsis of the various miR-185 dependent functional consequences is shown (Figure 2). More detailed information is provided as Supplementary Material for particular tumor entities, such as the sample types (tissue, plasma, cell lines, animal tissue) and numbers that were analyzed in the respective study as well as the observed molecular effects. In addition, information is given in the text paragraphs below about which target gene or pathway is known to be affected by miR-185-5p or miR-185-3p, respectively.

2. Oral and Pharyngeal Cancers

2.1. Biology

There is a growing body of research about the roles of miR-185 in the pathogenesis of oral and pharyngeal cancers (Supplementary Tables S1 and S2). Generally, miR-185 is downregulated in these tumors and acts as a tumor suppressor. Low expression of miR-185-3p in nasopharyngeal carcinoma patients is correlated with poor overall and recurrence-free survival as well as a weak response to radiotherapy by suppressing SMAD7 and WNT2B expression [16,17]. miR-185-3p also inhibits cellular growth and metastasis and promotes apoptosis in nasopharyngeal carcinoma cell lines [17,18]. SMAD7 functions as an antagonist of transforming growth factor β (TGF-β) type I receptor (TGF-βR1) and contributes to the development of various cancers [19]. High miR-185 expression induces apoptosis and autophagy via Homeobox C6 (HOXC6) transcription factor, repressing the oncogenic TGF-β1/mTOR pathway [20]. Low miR-185 expression and high levels of HOXC6 were found to be associated with lymph node metastasis, higher stages and lower survival. Variations of HOXC6, SMAD7, YWHAZ, FOXD3, RAB14, and ZNF703 exhibited specificity for oral cancers. However, there are no detailed studies about the diagnostic or prognostic value of miR-185 variations.
Enforced expression of miR-185 has reduced dysplasia, cell proliferation and angiogenesis in oral potentially malignant disorders (OPMDs) [21]. There was no significantly different miR-185-5p expression in tonsillar squamous cell carcinoma (TSCC), base of tongue squamous cell carcinoma (BOTSCC) and normal tonsillar tissue. However, high miR-185-5p expression was correlated to human papillomavirus (HPV)-negativity and decreased survival in TSCC/BOTSCC [22]. This implies that miR-185-5p may be more significant for tumor progression than carcinogenesis. In contrast to the above reports, miR-185 up- rather than downregulation in oral squamous cell carcinoma (OSCC) samples was reported by Ramdas et al. [23]. However, this contradictory data is based only on the very low number of just five investigated samples and needs to be considered with much caution.
Besides its involvement in tumorigenesis, the role of miR-185-5p in chemotherapy resistance was investigated. Its upregulation reduced cell viability in cisplatin-resistant tongue and larynx squamous cell carcinoma cell lines by downregulating the genes for aquaporin-3 (AQP3), caspase-14 (CASP-14) and arachidonate 12R-lipoxygenase (ALOX12B) [24]. More recently, miR-185 has been reported to considerably weaken OPMDs by reducing inflammation and inducing apoptosis through Akt/NF-kB and Akt/CASP-9 related pathways. In addition, while no significant difference in the miR-185-5p serum levels was found in oropharyngeal cancer patients and healthy subjects, serum levels were increased following radiotherapy and allowed an accurate prediction of severe radiation-induced xerostomia [25].

2.2. Regulation

Long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) are major upstream regulators of miR-185 (Figure 3). Higher levels of lncRNA Forkhead box D3 antisense RNA1 (FOXD3-AS1) led to increased expression of FOXD3 and the development of nasopharyngeal carcinoma through absorption of miR-185-3p [26]. Conversely, overexpression of miR-185-3p in nasopharyngeal carcinoma cells reversed oncogenic activities of FOXD3-AS1. circRNA circ0058106 suppressed miR-185-3p in hypopharyngeal squamous cell carcinoma thereby increasing proliferation, metastasis and EMT by activating the Wnt2b/β-catenin/c-Myc pathway [27]. Third, lncRNA LINC00958 promoted OSCC through sponging miR-185-5p [28]. Its overexpression directly suppressed miR-185-5p and indirectly enhanced YWHAZ expression, which is known to function as an oncogene [29]. Forced expression of miR-185-5p, however, counteracted this effect and suppressed YWHAZ both at mRNA and protein levels. lncRNA PDIA3P promoted OSCC progression through sponging miR-185-5p, thereafter activating CCND2 (cyclin D2). Interestingly, the CCND2 mRNA level was significantly higher in OSCC tissues, but its protein level was downregulated by miR-185-5p overexpression [30]. As a fifth molecules, lncRNA LSINCT5 was aberrantly upregulated in OSCC and drove tumor progression by decreasing miR-185-5p and increasing ZNF703 levels [31]; ZNF703 has been characterized to be an oncogene in OSCC and a target of miR-185-5p. Finally, lncRNA KCNQ1OT1 accelerated OSCC tumorigenesis and reduced apoptosis by direct inhibition of miR-185-5p [32].

3. Esophageal Cancer

3.1. Biology

There are inconsistent reports about the function of miR-185 in esophageal cancers (Supplementary Table S3). Some evidence shows that miR-185 is upregulated in esophageal squamous cell carcinoma (ESCC) patients and associated with tumor node metastasis (TNM) stages [33,34,35]; this may have diagnostic potential. On the other hand, high expression of miR-185 led to a favorable prognosis in esophageal carcinoma patients and suppressed the malignancy and growth of esophageal carcinoma cells in vivo [36]. Also, upregulation of miR-185 in ESCC cells reduced proliferation, invasion, migration and metastasis through downregulation of the genes of the receptor for advanced-glycation end products (RAGE) and transcription factor Six1 [37,38]. RAGE activates several signaling pathways in various cancers and is involved in processes, such as cell proliferation, autophagy and apoptosis [39]. The homeobox protein Six1 plays an oncogenic role in various cancers [40]. miR-185 has also been implicated in the radioresistance of esophageal cancer. In two studies, Su et al. [41] and Zheng et al. [42] produced miR profiles of radioresistant esophageal cancer cells and reported downregulation of miR-185 expression compared to the radiosensitive parental cell line. High plasma levels of miR-185 were also observed in ESCC patients who responded to radiotherapy [43].

3.2. Regulation

lncRNA that is highly expressed in hepatocellular carcinoma exhibits oncogenic activities in esophageal carcinoma by upregulating the kallikrein-related peptidase 5 (KLK5) gene through sponging miR-185 [36]. Liu and colleagues [44] observed that the lncRNA KLF3-AS1, which exhibits a low expression in tumor tissues, functions as a competing endogenous RNA (ceRNA) for miR-185-5p in ESCC. The Krüppel-like factor 3 (KLF3) gene exhibits a variety of biological effects, such as regulation of differentiation and apoptosis. The results suggest that KLF3 acts as a tumor suppressor in ESCC by inhibiting migration and invasion of tumor cells. Loss of KLF3-AS1 results in high levels of miR-185-5p, which in turn lead to KLF3 downregulation (Figure 3).

4. Gastric Cancer

4.1. Biology

Generally, miR-185-5p is downregulated in gastric cancer (GC) promoting cell proliferation and invasion while simultaneously inhibiting apoptosis and necrosis [45] (Supplementary Table S4). Several studies suggest a miR-185-dependent mechanism for regulating the antitumor effects of GKN1. Gastrokine 1 (GKN1) is a protein expressed in gastric mucosa and exhibits gastroprotective effects [46,47]. The expression levels of GNK1 and miR-185 are low in GC patients and negatively correlated with the methylation status of the CpG island methylator phenotype and high transcript levels of both the DNA methyltransferase 1 (DNMT1) and the enhancer of zeste homolog 2 (EZH2) gene [48]. miR-185 is induced by GKN1 and required for GKN1’s tumor-suppressive activities [49]. Interestingly, GKN1 increased the expression of miR-185 in GC cell lines by suppressing c-Myc, which was recognized as a suppressor of miR-185 by binding to its promoter. Increased miR-185 expression in GC cell lines led to cell cycle arrest and reduced proliferation by inactivating DNMT1 and EZH2 and increasing the expression of the cyclin dependent kinase inhibitor 2A (CDKN2A) and E-cadherin (CDH1) genes. The increased expression was the result of GKN1/miR-185/DNMT1/EZH2–mediated hypomethylation of the promoter regions. The N-terminal hydrophobic region and BRICHOS domain of GKN1 were sufficient for its tumor-suppressive functions [50]. Downregulated GKN1 and miR-185 have also been shown to be negatively correlated with the high expression of ras homolog gene family member A (RHOA) [51]. GKN1 promotes the miR-185 expression level in GC cell lines and significantly inhibits cell migration and invasion by repressing RHOA in a miR-185 dependent manner. RhoA is a member of the Rho subgroup of the Ras superfamily and exhibits oncogenic activities [52].
Some studies have reported that miR-185 is involved in regulating the chemosensitivity of GC. miR-185 is downregulated in drug-resistant GC cells. miR-185 inhibition increased resistance to chemotherapeutic drugs by upregulating multidrug resistance (MDR) genes [53]. Inversely, enhanced expression led to more chemosensitivity of GC cells by reducing the expression of apoptosis repressor with caspase recruitment domain (ARC) both in in vitro and in vivo [54]. An upregulation of miR-185 upon cisplatin or doxorubicin therapy was the result of direct binding of Runt-related transcription factor 3 (RUNX3) to the binding site 3 (BS3) region of the miR-185 promoter, which finally triggered the promoter activity of miR-185. Tan et al. [55] reported the involvement of miR-185 in zinc finger protein 139 (ZNF139) induced enhancement of MDR in GC cells. ZNF139 was confirmed to be an upstream regulator of miR-185 that diminishes miR-185 transcription. This increased MDR characteristics in GC cells through the induction of the MDR-associated genes MDR1/P-gp, MRP and BCL-2. In another study, a significantly lower level of serum miR-185 was observed in patients with advanced GC, but it increased after neoadjuvant chemotherapy with the SOX regimen [56]. This could be used as a biomarker to predict the outcomes of neoadjuvant chemotherapy.
miR-185 levels correlate with advanced clinical stage, lymph node metastasis and poor clinical outcomes [57]. Additionally, miR-185-5p expression was associated with clinicopathological characteristics, including tumor size, differentiation, and lymphatic metastasis [58]. Enforced expression suppressed the genes DNMT1 and tripartite motif-containing 29 (TRIM29) and resulted in a rescue of GC proliferation and metastasis, as well as an induction of apoptosis and cell cycle arrest [59]. TRIM29 is a transcription factor that has shown oncogenic or tumor-suppressive activities dependent on the tumor type. One of the mechanisms of the tumor suppressive activity of miR-185 is related to its ability to directly suppress the TRIM29-Wnt/β-catenin signaling axis. Interestingly, miR-185-5p mimics significantly increased apoptosis of GC cells by decreasing expression of BCL-2 and the gene of the X-linked inhibitor of apoptosis protein (XIAP), simultaneously increasing CASP-8 and CASP-3 expression and activities [58]. Enforced overexpression of miR-185-3p in GC cells led to a strong inhibition of EMT and migration and increased apoptosis through inactivation of the PI3K/Akt axis by downregulation of the cathepsin D gene (CTSD) [60].
Conversely to the data above, some studies reported high miR-185 levels in GC (Supplementary Table S4). Yao et al. [61], Treece et al. [62], and Zhang et al. [63] found considerable upregulation of miR-185 in GC patients, for example. Looking for biomarkers for GC detection, Zhou et al. [64] investigated the levels of miRs in plasma, GC tissues and plasma exosomes derived from GC patients. They observed an increased level of miR-185 in all three sample types. In addition, a higher level was detected in the plasma of patients with high- as compared to low-stage GC. In a similar study, miR-185-5p expression was found to be high in serum, tissue, and serum exosomes and, along with other miRs, showed diagnostic value for GC detection [65]. High miR-185 levels have also been associated with chemoresistance [66].

4.2. Regulation

In addition to lncRNAs, the genes GNK1, c-Myc, ZNF139, and RUNX3 encode specific upstream regulators of miR-185 in GC. Interestingly, other lncRNAs were identified to regulate the expression of miR-185 in GC than in oral, pharyngeal or esophageal cancers (Figure 3). lncRNA XIST has been shown to regulate miR-185 expression [67] and functioned as a ceRNA for miR-185 to decrease miR-185-induced TGF-β1 suppression and accelerate GC progression. Wu and colleagues [68] reported that lncRNA FOXD2-AS1 was able to increase apatinib resistance by direct suppression of miR-185-5p and decreased the inhibitory effects of miR-185-5p on CCND2. By contrast, ectopic expression of miR-185-5p could suppress the FOXD2-AS1 effect on apatinib resistance and sensitize GC cells to apatinib through CCND2 suppression [68].

5. Liver Cancer

5.1. Biology

There is growing evidence that miR-185 expression in the liver has implications through regulating several oncogenes in hepatocellular carcinoma (HCC) (Supplementary Table S5). This includes the gene cell division cycle 42 (CDC42) which belongs to the Rho GTPase family; its oncogenic activities are well documented in various cancers [69]. The Rho-associated coiled-coil containing protein kinase 2 gene (ROCK2) encodes for a serine/tyrosine kinase that is involved in controlling HCC proliferation, metastasis and chemoresistance [70]. CDC42 and ROCK2 are direct targets of miR-185 and overexpressed in HCC cells due to miR-185 deficiency [71,72]. When miR-185-5p expression was restored, cell migration and invasion capacity were dramatically reduced. A low level of miR-185-5p expression in HCC tissues was also positively correlated with aggressive clinicopathological features, such as high TNM stage and lymph node metastasis.
Integrin-β5 (ITGB5) is involved in the cell interaction with the extracellular matrix (ECM) and promotes HCC carcinogenesis by activation of the WNT/β-catenin pathway; homeobox gene SIX2 is a known oncogene. Both are targeted by miR-185 [73,74]. Through the inactivation of the WNT/β-catenin pathway in an ITGB5-dependent manner and the suppression of SIX2, miR-185 exerts anti-growth and anti-metastatic effects on HCC cells and reverses EMT. Qadir et al. [75] observed a low miR-185 expression in HCC tissues infected with hepatitis B (HBV) and hepatitis C viruses (HCV) as well as three different HCC cell lines. This study indicated that restoration of miR-185 expression suppressed DNMT1 expression and PTEN promoter methylation, increased PTEN expression and consequently PTEN-induced Akt inhibition and thus prevented HCC growth. Epigenetic control of PTEN by miR-185 has also been shown in cholangiocarcinoma [76]. Additionally, miR-185 was found to inhibit proliferation and cell cycle progression and induce apoptosis and autophagy in HCC cells by targeting different genes in the Akt signaling pathway, including AKT1, RICTOR and RHEB [77].
Downregulation of miR-185 is considered a prognostic marker of HCC. Zhi and colleagues [78] reported accurate prognosis of survival and tumor recurrence in patients with early-stage HCC, a finding that was confirmed by others [79]. Furthermore, miR-185 mimics caused a considerable suppression of cell growth and invasion in HCC cell lines. Serum miR profiling in HCV-positive HCC patients recognized six dysregulated miRs, which showed a high diagnostic accuracy for discriminating HCV-positive HCC from non-HCC individuals [80]. miR-185-5p was one of them, exhibiting lower abundance in cancer patients.
As opposed to the above-mentioned publications, a few studies have reported upregulation of miR-185 and an oncogenic activity in HCC (Supplementary Table S5). Wen and colleagues [81] evaluated the miR profiles of plasma derived from HBV-positive HCC patients. They highlighted eight overexpressed miRs, including miR-185-5p, with diagnostic potential. Likewise, Tang and colleagues [82] determined miR profiles in HCC clinical samples and found 20 miRs that were related to venous metastasis, again including miR-185. Placenta-specific 8 (PLAC8) or Onzin is a highly conserved cysteine-rich protein that is downregulated in HCC and acts as a tumor suppressor. High expression of miR-185-5p actually downregulates PLAC8 expression [83].

5.2. Regulation

Some lncRNAs have been shown to be involved in miR-185 regulation (Figure 3). One molecule is lncRNA FOXD2-AS1. Its knockdown effectively increased miR-185 expression, thus suppressing AKT and subsequently reducing proliferation, invasion and migration [84]. Linc00176 also seems to play an oncogenic role. Upon its knockdown, there is a release of miR-185-5p and an induction of cell cycle arrest and necroptosis [85]. LncRNA MEG3 is downregulated in HCC due to methylation of its promoter region. It exerts tumor-suppressive activities via regulation of p53 expression [86]. Zamani et al. [87] reported that dendrosomal curcumin increases miR-185 expression in HCC cells and thereby induces promoter DNA hypomethylation and upregulation of MEG3 through DNMT1 targeting.
Besides lncRNA molecules, miR-185 is regulated by a different mechanism via the receptor for activated protein kinase C (RACK1) [88]. RACK1 is downregulated in HCC tissues. While it is not needed for maturation, it is required for full miR-185 functioning. RACK1 interacts with a part of the Dicer complex called KH-type splicing regulatory protein (KSRP) and is necessary for the recruitment of mature miR-185 to RISC. Knockdown of RACK1 caused a weaker miR-185-mediated inhibition of the expression of target genes in HCC cells. Another process involves ELK1. It is a transcription factor that belongs to the ternary complex factor subfamily of the ETS family and is involved in viral life cycles and virus-associated diseases. Fan et al. [89] found that miR-185-5p inhibits HBV replication and gene expression in HCC cell lines in an ELK1-dependent manner and also reduced HBV preS1 promoter activity by inhibiting ELK1. A rescued ELK1 expression reversed the suppressive effects of miR-185-5p.

6. Pancreatic Cancer

Biology and Regulation

Pancreatic cancer (PDAC) mortality is close to incidence and will become the second most frequent cause of cancer-related death by about 2030. Concerning miR-185 (Supplementary Table S6), an inverse association between lncRNA XIST and miR-185-5p has been described. Enforced expression of miR-185-5p or knockdown of XIST in PDAC cell lines reduced cell proliferation and triggered cell cycle arrest and apoptosis. XIST also suppressed miR-185-5p through direct interaction, which in turn inhibited oncogene CCND2 [90]. lncRNA PCAT6 promotes progression and tumorigenesis of PDAC through sponging of miR-185-5p and a resulting upregulation of the chromobox 2 (CBX2) gene [91]. In contrast, miR-185-5p mimics interfered with oncogenic activities of PCAT6. Overexpression and oncogenic activities of CBX2 have also been reported in several other cancers. In another study, increased abundance of transcriptional coactivator with PDZ-binding motif (TAZ) has been shown in tissues, serum and pancreatic fluid of PDAC patients and is negatively correlated with miR-185 expression. TAZ is a 14-3-3 binding protein with confirmed oncogenic activities in PDAC [92]. miR-185 inhibited the proliferation of PDAC cells by downregulating the TAZ-encoding gene TAFAZZIN through the direct targeting of the 3′-UTR of its mRNA [93]. Rather than suppressing tumor-related activities, Gao et al. [94] reported that upregulation of miR-185 contributes to PDAC development by downregulation of NTRK3 and CORO2B. The analysis was based on data obtained from only few tissues, however. NTRK3 belongs to the neurotrophin receptor family and modulates cell survival, while CORO2B is an actin-binding protein. Diagnostically, higher levels of miR-185 and six other miRs (miR-20a, miR-21, miR-24, miR-25, miR-99a, and miR-191) were found in a comparison of PDAC patients to healthy donors and patients with chronic pancreatitis [95]. The panel could discriminate different tumor stages with high sensitivity and specificity. Overall, however, miR-185 has not been found to contribute significantly to blood-based diagnosis of PDAC [96].

7. Colorectal Cancer

7.1. Biology

Colorectal cancer (CRC) is the third most common cancer in the world, with a high mortality rate [97]. The role of miR-185 in CRC has been widely evaluated. Most studies confirm that miR-185 is downregulated and exhibits tumor suppressing activity via several cellular processes (Supplementary Table S7). The Wnt/β-catenin pathway is one of the targets implicated in CRC development. miR-185 upregulation reduced aggressive features of CRC by directly targeting WNT1 as well as MYC and CCND1 further downstream [98,99]. Urothelial carcinoma-associated 1 (UCA1) is an lncRNA that is significantly upregulated in CRC and involved in its progression [100]. UCA1 functions as a sponge for miR-185-5p, thereby activating the WNT1-inducible signaling pathway protein 2 (WISP2)/β-catenin pathway and promoting CRC.
Other studies have shown that miR-185-5p and miR-185-3p considerably reduced CRC growth and its metastatic and angiogenic potential by directly suppressing RHOA [101], CDC42 [101], c-MYC [102], and the gene of aquaporin 5 (AQP5) [103]. The stromal interaction molecule 1 (STIM1) gene is also directly affected [104]; it is an endoplasmic reticulum calcium sensor that activates the store-operated calcium influx when ER discharges calcium. Further, insulin-like growth factor 1 receptor (IGF1R) and insulin-like growth factor 2 (IGF2) are targeted by miR-185 [105]. IGF1R is a tyrosine kinase receptor that plays a pivotal role in angiogenesis, growth, metastasis and resistance to apoptosis in CRC. IGF2 triggers the signaling pathway associated with proliferation and survival of CRC through IGF1R. Hypoxia-inducible factor-2α (HIF-2α) is another verified target of miR-185 [106]. It increases hypoxic tumor cell proliferation through c-Myc activation and leads to CRC progression by deregulating iron homeostasis. Additionally, suppression of these genes by miR-185 noticeably inhibited EMT, increased apoptosis and reduced CRC resistance to chemotherapy and radiotherapy [103,104,105].
Yuan and colleagues [107] reported that miR-185 blocks MMP-9 and VEGF expression as well as the metastatic ability of CRC cells by repressing DC-SIGN. DC-SIGN is a C-type lectins domain family 4 member and abundantly expressed in immature dendritic cells. The miR-185 repression of DC-SIGN prevents β-catenin translocation to the nucleus of CRC cells by inactivation of the PI3K/Akt/GSK-3β pathway and the DC-SIGN/TCF1/LEF1 pathway could directly downregulate miR-185 in CRC. Li et al. [108] found a low-frequency 3′-UTR variant rs12915554 in GREM1—a member of the TGF-β superfamily—associated with enhanced CRC susceptibility. Interestingly, this variation stabilized the GREM1 transcript by disturbing the binding of miR-185-3p in CRC cells. In another paper, it was reported that miR-185-3p is downregulated in CRC due to the impairment of argonaute 2 (AGO2), a key regulator in miR processing [109]. Treatment with a miR-185-3p mimic could reduce metastasis by downregulating NRP1. NRP1 has been shown to interact with different ligands and receptors to increase tumorigenesis and EMT [110].
miR-185 expression has shown potential in diagnosis and prognosis of CRC. Significantly lower miR-185 expression was found in cancerous than non-cancerous tissues [111,112]. In addition, this was associated with advanced clinical stage and metastasis. Downregulation was also reported in CRC patients with subsequent relapse. miR-185 had a prognostic value for metastasis-free survival in combination with four other miRs [113]. Similarly, the miR-185 abundance in exosomes derived from serum was reduced in samples from patients with recurrence of liver metastasis [114]. The expression of miR-185 in circulating tumor cells derived from metastatic CRC patients during a chemotherapy course was transient and fluctuating [115]. Low miR-185-5p expression in CRC tissue samples is also associated with liver metastasis [116], while higher plasma levels were predictive for chemotherapy response. Expressions of miR-185 and that of affected genes, such as glutathione peroxidases 2 (GPX2) and selenophosphate synthetase 2 (SEPHS2), decreased considerably in tumor cells grown in selenium-deficient medium [117].
In contrast to the majority of findings, there are few studies that reported miR-185 upregulation in CRC, actually promoting carcinogenesis (Supplementary Table S7). Akcakaya et al. [118] showed correlation with poor survival and development of metastatic disease in CRC patients, implying that miR-185 might have a negative prognostic role in CRC. Recently, another study identified that miR-185-5p-induced suppression of AT-rich interaction domain 1A (ARID1A), a tumor suppressor gene, was associated with poor prognosis and adverse outcomes [119]. Zhang et al. [120], finally, reported that miR-185 was expressed at a high level in colon cancer stem cells and seems to be important in maintaining stemness. Moreover, its expression was significantly increased during the transformation of normal intestine treated with a carcinogenic agent [121].

7.2. Regulation

As for other types of digestive tract cancers, lncRNAs have been identified to trigger CRC progression via miR-185 regulation (Figure 3). High expression of lncRNA UCA1 in CRC cells led to a notable increase in the migration, invasion and EMT by binding to miR-185-5p, which directly activated the MAPK14/MAPKAPK2/Hsp27 axis [122] and repressed NOTCH3 [123]. In another study, overexpressed LINC00152 promoted CRC growth by sponging miR-185-3p and upregulating its direct target fascin actin-bundling protein 1 (FSCN1). However, miR-185-3p activation partially reversed this effect [124]. NEAT1 increased CRC metastasis through miR-185-5p absorption and IGF2 induction. By contrast, miR-185-5p mimics decreased NEAT1-induced CRC metastasis [125]. As shown also in HCC, FOXD2-AS1 downregulated miR-185-5p and thus upregulated its target CDC42. This resulted in an increased proliferation, migration and invasion capacity of CRC cells [126]. In turn, miR-185-5p overexpression considerably reversed the aggressive features. HAGLR is a recently discovered lncRNA that is aberrantly expressed in many malignancies. Recently, it was reported that HAGLR is upregulated in CRC and promotes cancer in a xenograft model by targeting miR-185-5p, leading to upregulated CDK4 and CDK6 [127]. This effect could be reversed by overexpressing miR-185-5p. Another study showed that high expression of circRNA ArfGAP with FG repeats 1 (circAGFG1) increased the expression of transcription factor YY1, induced CTNNB1 (β-catenin) and triggered metastasis and stemness by adsorbing miR-185-5p [128]. LncRNA differentiation antagonizing non-coding RNA (DANCR) has been shown to be upregulated in CRC [129]. Overexpression increases high mobility group A2 (HMGA2) expression by repressing miR-185-5p and promotes CRC progression. ASB16 antisense RNA 1 (ASB16-AS1) has also been shown to drive CRC progression by downregulating miR-185-5p and upregulating TEA domain transcription factor1 (TEAD1) as the main effector of Hippo signaling [130].

8. Conclusions

Dysregulation of miR-185 is strongly implicated in the pathogenesis of digestive tract cancers and associated with clinical outcome. The fact that the molecule is relevant to all cancers indicates that it has a central role in regulating tumor biology. Both miR-185-5p and miR-185-3p are functionally active in the various tumors and numerous target genes or pathways have been reported (Table 1). On the basis of the currently available data, it seems as if more genes are affected by miR-185-5p than miR-185-3p. However, this could be accidental or due to a bias in the studies performed. In addition, quite a few analyses did not specify the variant and only reported a role of miR-185 globally. It is reassuring, however, that no gene was found to be regulated by both miR-185-5p and miR-185-3p in different tumor entities. For three genes—CDC42, PLAC8, and RHOA—for which no information about the variant is available in some tumor types, comparison between different datasets or tumor entities may suggest which miR-185 version is responsible for their regulation. For these and all others, however, comparison to the respective target mRNA would provide the relevant information.
The fact that most regulation seemingly occurs via miR-185-5p and less via miR-185-3p suggests that oncogenic and tumor suppressive functions could be linked to one of the two mature miRs, respectively. Alternatively, the use of either miR-185 variant could be organ-related. However, no such correlation was apparent from the available data.
While many functional aspects of miR-185 variation have been demonstrated, the reported analyses have been focusing on particular gene functions, providing only many fragmented views while lacking a more comprehensive understanding of parallel or synergistic regulative processes and their interaction. A good example for this is the reports about the regulation of miR-185 in oral cancer and its functional consequences (Table 2). Each study looked at only one regulator of miR-185 and the effects that the reduced miR-185 level had for one or a few genes. While the same process—reduction of miR-185—was described in all cases, there was basically no apparent overlap between the results of the six studies.
About three quarters of the publications discussed in this review have reported that miR-185 predominantly exhibits tumor-suppressive functions. Conversely, however, some studies suggest that it functions as an oncomiR. Such contradictory results were reported for nearly all mentioned tumor forms. From the publications and data looked at as part of this review, there was no factor with apparent responsibility for this striking difference. However, the number of publications indicating an oncomiR function is too large to merely assume erroneous studies. In several cases, the relevant target genes responsible for the actual oncogenic activity have not been described. Further analyses are required to elucidate the reasons for the differences.
To some extent, the contradictory results might be associated with differences in methodologies and analyzed material. For example, Cao et al. [122] found that miR-185 was downregulated in CRC cell lines, SW480, SW620, and HT-29, compared to a non-cancerous colon cell line and miR-185 mimics inhibited cell proliferation and migration. Baldi et al. [119] reported instead overexpression of miR-185 in CRC cell lines HCT116 and LoVo and thereby oncogenic activities. Analyzing miR-185 expression in only relatively few samples may also result in contradictory results. Interestingly, however, there are results that were produced with the same methodology and on the same cell lines but nevertheless led to opposing results. Zou et al. [83] found that overexpression of miR-185 triggered oncogenic activities and enhanced cell viability in HCC cell lines Huh-7 and HepG2. By contrast, Tran et al. [85] showed that miR-185 inhibition rescued the same two HCC cell lines from cell death. One cannot exclude entirely that an undetected heterogeneity between the actual cell lines used in the different laboratories may be a factor, which would be difficult to unravel retrospectively, though.
In terms of biology, the contradictory roles of miR-185 may be dependent on its expression level and cellular abundance in relation to the expression of its targets (dose-dependent effects), as has been reported for other miRs [131]. Another explanation could be genomic heterogeneity. This is a known characteristic of human cancers and has been observed in tumors derived from different patients and even within a tumor from an individual patient [132]. Due to the context-dependent function of miRs [133], contextualization of miR-185 in specific genetic backgrounds may be an explanation for the functional discrepancies.
Several lncRNAs and circRNAs as well as AGO2, c-Myc, GNK1, RUNX3, RACK1, ZNF139, and the DC-SIGN/TCF1/LEF1 pathway have been reported as upstream regulators implicated in controlling miR-185 abundance and function in digestive tract cancers. Of these, lncRNA FOXD2-AS1 is an lncRNA that commonly regulates miR-185 abundance in colorectal, gastric, and liver cancers. Additionally, lncRNA XIST regulates miR-185 in both gastric and pancreatic cancers. However, identification of these overlapping processes in regulation is rather coincidental. Still missing to date are comprehensive studies of the mechanisms involved in the control and regulative processes of miR-185; synergies or complementation may have an effect that can only be revealed by such analyses. Moreover, epigenetic mechanisms, such as histone modifications and promoter DNA methylation, have not been investigated much.
In terms of functioning as a biomarker, several studies looked at the diagnostic and prognostic potential of miR-185 in tissue, plasma/serum or exosomes derived from patient blood. miR-185 is likely to be informative in combination with other features, acting more as a pan-cancer marker. However, there is still too little knowledge about the rules governing the changes and the processes that are possibly involved, such as an exosomal transfer of miR-185. Furthermore, the association between miR-185 expression and tumor microenvironment and immune cell infiltrations has not been examined yet. Particularly for CRC, miR-185 could even hold some promise as a therapeutic target, but much more data and detailed information will be required to assess its real potential.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ncrna8050067/s1, Table S1: miR-185 expression in oral cancers; Table S2: miR-185 expression in nasopharyngeal cancers; Table S3: miR-185 expression in esophageal cancers; Table S4: miR-185 expression in gastric cancers; Table S5: miR-185 expression in hepatocellular carcinoma (HCC); Table S6: miR-185 expression in pancreatic cancer (PDAC); Table S7: miR-185 expression in colorectal cancer (CRC).

Author Contributions

Conceptualization, E.B. and M.M.S.; methodology, E.B., F.N., M.R. and Z.J.; investigation, E.B., F.H.M., F.N., M.D.F., M.R., S.S. and Z.J.; data curation, F.H.M., M.D.F. and S.S.; writing—original draft preparation, E.B., J.D.H. and M.M.S.; writing—review and editing, E.B., J.D.H. and M.M.S.; project administration, E.B., J.D.H. and M.M.S. All authors have read and agreed to the published version of the manuscript.

Funding

Soroosh Shahryarhesami was supported by a fellowship of the Deutscher Akademischer Austauschdienst (DAAD; 57048249).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef]
  2. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  3. Orditura, M.; Galizia, G.; Sforza, V.; Gambardella, V.; Fabozzi, A.; Laterza, M.M.; Andreozzi, F.; Ventriglia, J.; Savastano, B.; Mabilia, A.; et al. Treatment of gastric cancer. World J. Gastroenterol. 2014, 20, 1635–1649. [Google Scholar] [CrossRef]
  4. Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Stenzinger, A.; Weichert, W. Genetic profiling of cancers of the digestive system: Biological insights and clinical implications. Pathobiology 2017, 84, 306–322. [Google Scholar] [CrossRef] [PubMed]
  6. Vedeld, H.M.; Goel, A.; Lind, G.E. Epigenetic biomarkers in gastrointestinal cancers: The current state and clinical perspectives. Semin. Cancer Biol. 2018, 51, 36–49. [Google Scholar] [CrossRef]
  7. Tazawa, H.; Nagasaka, T.; Kagawa, S.; Fujiwara, T. MicroRNA as a molecular target for gastrointestinal cancers. Transl. Gastrointest. Cancer 2015, 4, 219–235. [Google Scholar]
  8. Stang, A.; Weilert, H.; Lipp, M.J.; Oldhafer, K.J.; Hoheisel, J.D.; Zhang, C.; Bauer, A.S. MicroRNAs in blood act as biomarkers of colorectal cancer and indicate potential therapeutic targets. Mol. Oncol. 2021, 15, 2480–2490. [Google Scholar] [CrossRef] [PubMed]
  9. Botla, S.K.; Savant, S.; Jandaghi, P.; Bauer, A.S.; Mücke, O.; Moskalev, E.A.; Neoptolemos, J.P.; Costello, E.; Greenhalf, W.; Scarpa, A.; et al. Early epigenetic down-regulation of microRNA-192 expression promotes pancreatic cancer progression. Cancer Res. 2016, 76, 4149–4159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Dror, S.; Sander, L.; Schwartz, H.; Sheinboim, D.; Barzilai, A.; Dishon, Y.; Apcher, S.; Golan, T.; Greenberger, S.; Barshack, I.; et al. Melanoma miRNA trafficking controls tumour primary niche formation. Nat. Cell Biol. 2016, 18, 1006–1017. [Google Scholar] [CrossRef]
  11. Keller, A.; Leidinger, P.; Vogel, B.; Backes, C.; ElSharawy, A.; Galata, V.; Mueller, S.C.; Marquart, S.; Schrauder, M.G.; Strick, R.; et al. miRNAs can be generally associated with human pathologies as exemplified for miR-144. BMC Med. 2014, 12, 224. [Google Scholar] [CrossRef] [PubMed]
  12. Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef] [PubMed]
  13. Winter, J.; Jung, S.; Keller, S.; Gregory, R.I.; Diederichs, S. Many roads to maturity: Microrna biogenesis pathways and their regulation. Nat. Cell Biol. 2009, 11, 228–234. [Google Scholar] [CrossRef] [PubMed]
  14. Peng, Y.; Croce, C.M. The role of MicroRNAs in human cancer. Signal Transduct. Target. Ther. 2016, 1, 15004. [Google Scholar] [CrossRef] [Green Version]
  15. Galvão-Lima, L.J.; Morais, A.H.F.; Valentim, R.A.M.; Barreto, E.J.S.S. miRNAs as biomarkers for early cancer detection and their application in the development of new diagnostic tools. Biomed. Eng. Online 2021, 20, 21. [Google Scholar] [CrossRef]
  16. Li, G.; Wang, Y.; Liu, Y.; Su, Z.; Liu, C.; Ren, S.; Deng, T.; Huang, D.; Tian, Y.; Qiu, Y. miR-185-3p regulates nasopharyngeal carcinoma radioresistance by targeting WNT2B in vitro. Cancer Sci. 2014, 105, 1560–1568. [Google Scholar] [CrossRef] [Green Version]
  17. Xu, J.; Ai, Q.; Cao, H.; Liu, Q. MiR-185-3p and miR-324-3p predict radiosensitivity of nasopharyngeal carcinoma and modulate cancer cell growth and apoptosis by targeting SMAD7. Med. Sci. Monit. 2015, 21, 2828–2836. [Google Scholar] [CrossRef] [Green Version]
  18. Liu, C.; Li, G.; Ren, S.; Su, Z.; Wang, Y.; Tian, Y.; Liu, Y.; Qiu, Y. miR-185-3p regulates the invasion and metastasis of nasopharyngeal carcinoma by targeting WNT2B in vitro. Oncol. Lett. 2017, 13, 2631–2636. [Google Scholar] [CrossRef] [Green Version]
  19. de Ceuninck van Capelle, C.; Spit, M.; Ten Dijke, P. Current perspectives on inhibitory SMAD7 in health and disease. Crit. Rev. Biochem. Mol. Biol. 2020, 55, 691–715. [Google Scholar] [CrossRef]
  20. Cheng, J.-Z.; Chen, J.-J.; Wang, Z.-G.; Yu, D. MicroRNA-185 inhibits cell proliferation while promoting apoptosis and autophagy through negative regulation of TGF-β1/mTOR axis and HOXC6 in nasopharyngeal carcinoma. Cancer Biomark. 2018, 23, 107–123. [Google Scholar] [CrossRef]
  21. Wang, L.; Yin, P.; Wang, J.; Wang, Y.; Sun, Z.; Zhou, Y.; Guan, X. Delivery of mesenchymal stem cells-derived extracellular vesicles with enriched miR-185 inhibits progression of OPMD. Artif. Cells Nanomed. Biotechnol. 2019, 47, 2481–2491. [Google Scholar] [CrossRef] [PubMed]
  22. Bersani, C.; Mints, M.; Tertipis, N.; Haeggblom, L.; Näsman, A.; Romanitan, M.; Dalianis, T.; Ramqvist, T. MicroRNA-155, -185 and -193b as biomarkers in human papillomavirus positive and negative tonsillar and base of tongue squamous cell carcinoma. Oral Oncol. 2018, 82, 8–16. [Google Scholar] [CrossRef] [PubMed]
  23. Ramdas, L.; Giri, U.; Ashorn, C.L.; Coombes, K.R.; El-Naggar, A.; Ang, K.K.; Story, M.D. miRNA expression profiles in head and neck squamous cell carcinoma and adjacent normal tissue. Head Neck 2009, 31, 642–654. [Google Scholar] [CrossRef]
  24. Ratovitski, E.A. Phospho-ΔNp63α regulates AQP3, ALOX12B, CASP14 and CLDN1 expression through transcription and microRNA modulation. FEBS Lett. 2013, 587, 3581–3586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Tomasik, B.; Papis-Ubych, A.; Stawiski, K.; Fijuth, J.; Kędzierawski, P.; Sadowski, J.; Stando, R.; Bibik, R.; Graczyk, Ł.; Latusek, T.; et al. Serum microRNAs as xerostomia biomarkers in patients with oropharyngeal cancer undergoing radiation therapy. Int. J. Radiat. Oncol. Biol. Phys. 2021, 111, 1237–1249. [Google Scholar] [CrossRef] [PubMed]
  26. Hu, J.; Pan, J.; Luo, Z.; Duan, Q.; Wang, D. Long non-coding RNA FOXD3-AS1 silencing exerts tmor suppressive effects in nasopharyngeal carcinoma by downregulating FOXD3 expression via microRNA-185-3p upregulation. Cancer Gene Ther. 2021, 28, 602–618. [Google Scholar] [CrossRef]
  27. Li, C.; Li, W.; Cao, S.; Xu, J.; Qian, Y.; Pan, X.; Lei, D.; Wei, D. Circ_0058106 promotes proliferation, metastasis and EMT process by regulating Wnt2b/β-catenin/c-Myc pathway through miR-185-3p in hypopharyngeal squamous cell carcinoma. Cell Death Dis. 2021, 12, 1063. [Google Scholar] [CrossRef]
  28. Wang, Z.; Zhu, X.; Dong, P.; Cai, J. Long noncoding RNA LINC00958 promotes the oral squamous cell carcinoma by sponging miR-185-5p/YWHAZ. Life Sci. 2020, 242, 116782. [Google Scholar] [CrossRef]
  29. Gan, Y.; Ye, F.; He, X.X. The role of YWHAZ in cancer: A maze of opportunities and challenges. J. Cancer 2020, 11, 2252–2264. [Google Scholar] [CrossRef]
  30. Sun, C.-C.; Zhang, L.; Li, G.; Li, S.J.; Chen, Z.L.; Fu, Y.F.; Gong, F.Y.; Bai, T.; Zhang, D.Y.; Wu, Q.M.; et al. The lncRNA PDIA3P interacts with miR-185-5p to modulate oral squamous cell carcinoma progression by targeting cyclin D2. Mol. Ther. Nucleic Acid. 2017, 9, 100–110. [Google Scholar] [CrossRef] [Green Version]
  31. Wang, X.; Feng, X.; Wang, H. LncRNA LSINCT5 drives proliferation and migration of oral squamous cell carcinoma through the miRNA-185-5p/ZNF703 axis. J. Balk. Union Oncol. 2021, 26, 124–131. [Google Scholar]
  32. Bao, Q.; Liao, X.; Li, R.; Ding, N. KCNQ1OT1 promotes migration and inhibits apoptosis by modulating miR-185-5p/Rab14 axis in oral squamous cell carcinoma. Dev. Growth Differ. 2019, 61, 466–474. [Google Scholar] [CrossRef] [PubMed]
  33. Li, B.X.; Yu, Q.; Shi, Z.L.; Li, P.; Fu, S. Circulating microRNAs in esophageal squamous cell carcinoma: Association with locoregional staging and survival. Int. J. Clin. Exp. Med. 2015, 8, 7241–7250. [Google Scholar] [PubMed]
  34. Li, C.Y.; Zhang, W.W.; Xiang, J.L.; Wang, X.H.; Li, J.; Wang, J.L. Identification of microRNAs as novel biomarkers for esophageal squamous cell carcinoma: A study based on the Cancer Genome Atlas (TCGA) and bioinformatics. Chin. Med. J. 2019, 132, 2213–2222. [Google Scholar] [CrossRef] [PubMed]
  35. Yao, C.; Li, Y.; Luo, L.; Xiong, Q.; Zhong, X.; Xie, F.; Feng, P. Identification of miRNAs and genes for predicting Barrett’s esophagus progressing to esophageal adenocarcinoma using miRNA-mRNA integrated analysis. PLoS ONE 2021, 16, e0260353. [Google Scholar] [CrossRef]
  36. Wang, B.; Hao, X.; Li, X.; Liang, Y.; Li, F.; Yang, K.; Chen, H.; Lv, F.; Gao, Y. Long noncoding RNA HEIH depletion depresses esophageal carcinoma cell progression by upregulating microRNA-185 and downregulating KLK5. Cell Death Dis. 2020, 11, 1002. [Google Scholar] [CrossRef]
  37. Jing, R.; Chen, W.; Wang, H.; Ju, S.; Cong, H.; Sun, B.; Jin, Q.; Chu, S.; Xu, L.; Cui, M. Plasma miR-185 is decreased in patients with esophageal squamous cell carcinoma and might suppress tumor migration and invasion by targeting RAGE. Am. J. Physiol. Gastrointest. Liver Physiol. 2015, 309, G719–G729. [Google Scholar] [CrossRef] [Green Version]
  38. Zhao, Z.T.; Zhou, W.; Liu, L.Y.; Lan, T.; Zhan, Q.M.; Song, Y.M. Molecular mechanism and effect of microRNA185 on proliferation, migration and invasion of esophageal squamous cell carcinoma. Natl. Med. J. China 2013, 93, 1426–1431. [Google Scholar]
  39. Palanissami, G.; Paul, S.F.D. RAGE and Its Ligands: Molecular Interplay between Glycation, Inflammation, and Hallmarks of Cancer—A Review. Horm. Cancer 2018, 9, 295–325. [Google Scholar] [CrossRef]
  40. Wu, W.; Ren, Z.; Li, P.; Yu, D.; Chen, J.; Huang, R.; Liu, H. Six1: A critical transcription factor in tumorigenesis. Int. J. Cancer 2015, 136, 1245–1253. [Google Scholar] [CrossRef]
  41. Su, H.; Jin, X.; Zhang, X.; Xue, S.; Deng, X.; Shen, L.; Fang, Y.; Xie, C. Identification of microRNAs involved in the radioresistance of esophageal cancer cells. Cell Biol. Int. 2014, 38, 318–325. [Google Scholar] [CrossRef]
  42. Zheng, Z.F.; Su, H.F.; Zou, Y.; Peng, Z.; Wu, S.X. Expression profiles of microRNAs in radioresistant esophageal cell line. Natl. Med. J. China 2011, 91, 639–642. [Google Scholar]
  43. Martin, T.E.; Powell, C.T.; Wang, Z.; Bhattacharyya, S.; Walsh-Reitz, M.M.; Agarwal, K.; Toback, F.G. A novel mitogenic protein that is highly expressed in cells of the gastric antrum mucosa. Am. J. Physiol. Gastrointest. Liver Physiol. 2003, 285, G332–G343. [Google Scholar] [CrossRef] [Green Version]
  44. Liu, J.-Q.; Deng, M.; Xue, N.-N.; Li, T.X.; Guo, Y.X.; Gao, L.; Zhao, D.; Fan, R.T. LncRNA KLF3-AS1 suppresses cell migration and invasion in ESCC by impairing miR-185-5p-targeted KLF3 inhibition. Mol. Ther. Nucleic Acid. 2020, 20, 231–241. [Google Scholar] [CrossRef] [PubMed]
  45. Bibi, F.; Pushparaj, P.N.; Naseer, M.I.; Yasir, M.; Azhar, E.I. Decrypting the microRNA signatures in gastric cancer using high-throughput miRNA array coupled with systems biological approaches for precision medicine. Genet. Mol. Res. 2017, 16, 16039845. [Google Scholar] [CrossRef]
  46. Xing, R.; Li, W.; Cui, J.; Zhang, J.; Kang, B.; Wang, Y.; Wang, Z.; Liu, S.; Lu, Y. Gastrokine 1 induces senescence through p16/Rb pathway activation in gastric cancer cells. Gut 2012, 61, 43–52. [Google Scholar] [CrossRef]
  47. Alarcón-Millán, J.; Martínez-Carrillo, D.N.; Peralta-Zaragoza, O.; Fernández-Tilapa, G. Regulation of GKN1 expression in gastric carcinogenesis: A problem to resolve (Review). Int. J. Oncol. 2019, 55, 555–569. [Google Scholar] [PubMed]
  48. Choi, B.J.; Yoon, J.H.; Choi, W.S.; Kim, O.; Nam, S.W.; Lee, J.Y.; Park, W.S. GKN1 and miR-185 are associated with CpG island methylator phenotype in gastric cancers. Mol. Cell. Toxicol. 2013, 9, 227–233. [Google Scholar] [CrossRef]
  49. Yoon, J.H.; Choi, Y.J.; Choi, W.S.; Ashktorab, H.; Smoot, D.T.; Nam, S.W.; Lee, J.Y.; Park, W.S. GKN1-miR-185-DNMT1 axis suppresses gastric carcinogenesis through regulation of epigenetic alteration and cell cycle. Clin. Cancer Res. 2013, 19, 4599–4610. [Google Scholar] [CrossRef] [Green Version]
  50. Yoon, J.H.; Choi, Y.J.; Choi, W.S.; Nam, S.W.; Lee, J.Y.; Park, W.S. Functional analysis of the NH2-terminal hydrophobic region and BRICHOS domain of GKN1. Biochem. Biophys. Res. Commun. 2013, 440, 689–695. [Google Scholar] [CrossRef]
  51. Yoon, J.H.; Choi, W.S.; Kim, O.; Choi, B.J.; Nam, S.W.; Lee, J.Y.; Park, W.S. Gastrokine 1 inhibits gastric cancer cell migration and invasion by downregulating RhoA expression. Gastric Cancer 2017, 20, 274–285. [Google Scholar] [CrossRef] [Green Version]
  52. Hao, T.; Jiang, J.; Wu, W.; Tanaka, A.; Katoh, H.; Aburatani, H.; Lauwers, G.Y.; Fukayama, M. RhoA mutations in diffuse-type gastric cancer. Dig. Med. Res. 2020, 3, 4. [Google Scholar] [CrossRef]
  53. Li, Y.; Tan, B.B.; Zhao, Q.; Fan, L.Q.; Wang, D.; Liu, Y.; Liu, Q.W. Effect and mechanism of miR-185 inhibition to drug resistance of human gastric cancer cell line SGC7901. Chin. J. Cancer Prev. Treat. 2015, 22, 1347–1351. [Google Scholar]
  54. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Tan, B.; Li, Y.; Zhao, Q.; Fan, L.; Wang, D. ZNF139 increases multidrug resistance in gastric cancer cells by inhibiting miR-185. Biosci. Rep. 2018, 38, BSR20181023. [Google Scholar] [CrossRef]
  56. Tan, B.; Li, Y.; Di, Y.; Fan, L.; Zhao, Q.; Liu, Q.; Wang, D.; Jia, N. Clinical value of peripheral blood microRNA detection in evaluation of SOX regimen as neoadjuvant chemotherapy for gastric cancer. J. Clin. Lab. Anal. 2018, 32, e22363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Tan, Z.; Jiang, H.; Wu, Y.; Xie, L.; Dai, W.; Tang, H.; Tang, S. MiR-185 is an independent prognosis factor and suppresses tumor metastasis in gastric cancer. Mol. Cell. Biochem. 2014, 386, 223–231. [Google Scholar] [CrossRef] [PubMed]
  58. Fan, L.; Tan, B.; Li, Y.; Zhao, Q.; Yuan, H.; Liu, Y.; Wang, D.; Zhang, Z. Upregulation of miR-185 promotes apoptosis of the human gastric cancer cell line MGC803. Mol. Med. Rep. 2018, 17, 3115–3122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Qiu, F.; Xiong, J.-P.; Deng, J.; Xiang, X.-J. TRIM29 functions as an oncogene in gastric cancer and is regulated by miR-185. Int. J. Clin. Exp. Pathol. 2015, 8, 5053–5061. [Google Scholar] [PubMed]
  60. Huang, C.; Wu, Y.-J.; He, W.-F.; Zhao, S.L.; Ouyang, Y.Y.; Ai, X.H.; Liu, Z.Q.; Tang, S.Y. MiR-185-3p regulates epithelial mesenchymal transition via PI3K/Akt signaling pathway by targeting cathepsin D in gastric cancer cells. Transl. Cancer Res. 2020, 9, 6988–7000. [Google Scholar] [CrossRef] [PubMed]
  61. Yao, Y.; Suo, A.L.; Li, Z.F.; Liu, L.Y.; Tian, T.; Ni, L.; Zhang, W.G.; Nan, K.J.; Song, T.S.; Huang, C. MicroRNA profiling of human gastric cancer. Mol. Med. Rep. 2009, 2, 963–970. [Google Scholar] [PubMed] [Green Version]
  62. Treece, A.L.; Duncan, D.L.; Tang, W.; Elmore, S.; Morgan, D.R.; Dominguez, R.L.; Speck, O.; Meyers, M.O.; Gulley, M.L. Gastric adenocarcinoma microRNA profiles in fixed tissue and in plasma reveal cancer-associated and Epstein-Barr virus-related expression patterns. Lab. Investig. 2016, 96, 661–671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Zhang, F.; Xu, W.; Liu, J.; Liu, X.; Huo, B.; Li, B.; Wang, Z. Optimizing miRNA-module diagnostic biomarkers of gastric carcinoma via integrated network analysis. PLoS ONE 2018, 13, e0198445. [Google Scholar] [CrossRef] [Green Version]
  64. Zhou, X.; Zhu, W.; Li, H.; Wen, W.; Cheng, W.; Wang, F.; Wu, Y.; Qi, L.; Fan, Y.; Chen, Y.; et al. Diagnostic value of a plasma microRNA signature in gastric cancer: A microRNA expression analysis. Sci. Rep. 2015, 5, 11251. [Google Scholar] [CrossRef] [Green Version]
  65. Huang, Z.; Zhu, D.; Wu, L.; He, M.; Zhou, X.; Zhang, L.; Zhang, H.; Wang, W.; Zhu, J.; Cheng, W.; et al. Six serum-based miRNAs as potential diagnostic biomarkers for gastric cancer. Cancer Epidemiol. Biomark. Prev. 2017, 26, 188–196. [Google Scholar] [CrossRef] [PubMed]
  66. Kim, C.; Kim, H.K.; Rettig, R.L.; Kim, J.; Lee, E.T.; Aprelikova, O.; Choi, I.J.; Munroe, D.J.; Green, J.E. MiRNA signature associated with outcome of gastric cancer patients following chemotherapy. BMC Med. Genom. 2011, 4, 79. [Google Scholar] [CrossRef] [Green Version]
  67. Zhang, Q.; Chen, B.; Liu, P.; Yang, J. XIST promotes gastric cancer (GC) progression through TGF-β1 via targeting miR-185. J. Cell. Biochem. 2018, 119, 2787–2796. [Google Scholar] [CrossRef]
  68. Wu, X.; Shan, Y.; Cao, F.; Zhang, B.; Wang, H.; Liu, H.; Xu, Y.; Yu, H. Long noncoding RNA FOXD2-AS1 incuced gastric cancer cell MGC-803 to apatinib resistance by regulating miR-185-5p/CCND2 modulating axis. Chin. J. Cancer Biother. 2018, 25, 1104–1112. [Google Scholar]
  69. Arias-Romero, L.E.; Chernoff, J. Targeting Cdc42 in cancer. Expert Opin. Ther. Target. 2013, 17, 1263–1273. [Google Scholar] [CrossRef] [Green Version]
  70. Ma, W.; Sze, K.M.F.; Chan, L.K.; Lee, J.M.; Wei, L.L.; Wong, C.M.; Lee, T.K.; Wong, C.C.; Ng, I.O. RhoE/ROCK2 regulates chemoresistance through NF-κB/IL-6/STAT3 signaling in hepatocellular carcinoma. Oncotarget 2016, 7, 41445–41459. [Google Scholar] [CrossRef] [Green Version]
  71. Zhang, Q.; Chen, Y.; Liu, K. miR-185 inhibits cell migration and invasion of hepatocellular carcinoma through CDC42. Oncol. Lett. 2018, 16, 3101–3107. [Google Scholar] [CrossRef] [Green Version]
  72. Niu, Y.; Tang, G. miR-185-5p targets ROCK2 and inhibits cell migration and invasion of hepatocellular carcinoma. Oncol. Lett. 2019, 17, 5087–5093. [Google Scholar] [CrossRef] [Green Version]
  73. Lin, Z.; He, R.; Luo, H.; Lu, C.; Ning, Z.; Wu, Y.; Han, C.; Tan, G.; Wang, Z. Integrin-β5, a MIR-185-targeted gene, promotes hepatocellular carcinoma tumorigenesis by regulating β-catenin stability. J. Exp. Clin. Cancer Res. 2018, 37, 17. [Google Scholar] [CrossRef]
  74. Zhu, S.-M.; Chen, C.-M.; Jiang, Z.-Y.; Yuan, B.; Ji, M.; Wu, F.H.; Jin, J. MicroRNA-185 inhibits cell proliferation and epithelial-mesenchymal transition in hepatocellular carcinoma by targeting Six2. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 1712–1719. [Google Scholar] [PubMed]
  75. Qadir, X.V.; Han, C.; Lu, D.; Zhang, J.; Wu, T. MiR-185 inhibits hepatocellular carcinoma growth by targeting the DNMT1/PTEN/Akt pathway. Am. J. Pathol. 2014, 184, 2355–2364. [Google Scholar] [CrossRef] [Green Version]
  76. Qadir, X.V.; Han, C.; Wu, T. Epigenetic regulation of PTEN by miR-185 and miR-200b in liver cancer. Am. Assoc. Cancer Res. 2012, 78, 3143. [Google Scholar] [CrossRef]
  77. Zhou, L.; Liu, S.; Han, M.; Feng, S.; Liang, J.; Li, Z.; Li, Y.; Lu, H.; Liu, T.; Ma, Y.; et al. MicroRNA-185 induces potent autophagy via AKT signaling in hepatocellular carcinoma. Tumor Biol. 2017, 39, 1010428317694313. [Google Scholar] [CrossRef] [Green Version]
  78. Zhi, Q.; Zhu, J.; Guo, X.; He, S.; Xue, X.; Zhou, J.; Hu, B.; Li, H.; Chen, S.; Zhao, H.; et al. Metastasis-related miR-185 is a potential prognostic biomarker for hepatocellular carcinoma in early stage. Biomed. Pharmacother. 2013, 67, 393–398. [Google Scholar] [CrossRef]
  79. Kim, M.; Kogan, N.; Slack, F.J. Cis-acting elements in its 3′ UTR mediate post-transcriptional regulation of KRAS. Oncotarget 2016, 7, 11770–11784. [Google Scholar] [CrossRef] [Green Version]
  80. Li, J.; Jin, B.; Wang, T.; Li, W.; Wang, Z.; Zhang, H.; Song, Y.; Li, N. Serum microRNA expression profiling identifies serum biomarkers for HCV-related hepatocellular carcinoma. Cancer Biomark. 2019, 26, 501–512. [Google Scholar] [CrossRef] [PubMed]
  81. Wen, Y.; Han, J.; Chen, J.; Dong, J.; Xia, Y.; Liu, J.; Jiang, Y.; Dai, J.; Lu, J.; Jin, G.; et al. Plasma miRNAs as early biomarkers for detecting hepatocellular carcinoma. Int. J. Cancer 2015, 137, 1679–1690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Budhu, A.; Jia, H.L.; Forgues, M.; Liu, C.G.; Goldstein, D.; Lam, A.; Zanetti, K.A.; Ye, Q.H.; Qin, L.X.; Croce, C.M.; et al. Identification of metastasis-related microRNAs in hepatocellular carcinoma. Hepatology 2008, 47, 897–907. [Google Scholar] [CrossRef]
  83. Zou, L.; Chai, J.; Gao, Y.; Guan, J.; Liu, Q.; Du, J.J. Down-regulated PLAC8 promotes hepatocellular carcinoma cell proliferation by enhancing PI3K/Akt/GSK3β/Wnt/β-catenin signaling. Biomed. Pharmacother. 2016, 84, 139–146. [Google Scholar] [CrossRef] [PubMed]
  84. Chen, Z.; Zhang, Z.; Zhao, D.; Feng, W.; Meng, F.; Han, S.; Lin, B.; Shi, X. Long Noncoding RNA (lncRNA) FOXD2-AS1 promotes cell proliferation and metastasis in hepatocellular carcinoma by regulating MiR-185/AKT Axis. Med. Sci. Monit. 2019, 25, 9618–9629. [Google Scholar] [CrossRef] [PubMed]
  85. Tran, D.D.H.; Kessler, C.; Niehus, S.E.; Mahnkopf, M.; Koch, A.; Tamura, T. Myc target gene, long intergenic noncoding RNA, Linc00176 in hepatocellular carcinoma regulates cell cycle and cell survival by titrating tumor suppressor microRNAs. Oncogene 2018, 37, 75–85. [Google Scholar] [CrossRef]
  86. Braconi, C.; Kogure, T.; Valeri, N.; Huang, N.; Nuovo, G.; Costinean, S.; Negrini, M.; Miotto, E.; Croce, C.M.; Patel, T. MicroRNA-29 can regulate expression of the long non-coding RNA gene MEG3 in hepatocellular cancer. Oncogene 2011, 30, 4750–4756. [Google Scholar] [CrossRef]
  87. Zamani, M.; Sadeghizadeh, M.; Behmanesh, M.; Najafi, F. Dendrosomal curcumin increases expression of the long non-coding RNA gene MEG3 via up-regulation of epi-miRs in hepatocellular cancer. Phytomedicine 2015, 22, 961–967. [Google Scholar] [CrossRef]
  88. Otsuka, M.; Takata, A.; Yoshikawa, T.; Kojima, K.; Kishikawa, T.; Shibata, C.; Takekawa, M.; Yoshida, H.; Omata, M.; Koike, K. Receptor for activated protein kinase C: Requirement for efficient microRNA function and reduced expression in hepatocellular carcinoma. PLoS ONE 2011, 6, e24359. [Google Scholar] [CrossRef]
  89. Fan, H.X.; Feng, Y.J.; Zhao, X.P.; He, Y.Z.; Tang, H. MiR-185-5p suppresses HBV gene expression by targeting ELK1 in hepatoma carcinoma cells. Life Sci. 2018, 213, 9–17. [Google Scholar] [CrossRef]
  90. Wang, Y.-P.; Huang, Y.; Hou, T.; Lu, M. LncRNA XIST acts as a ceRNA sponging miR-185-5p to modulate pancreatic cancer cell proliferation via targeting CCND2. Transl. Cancer Res. 2020, 9, 1427–1438. [Google Scholar] [CrossRef]
  91. Wang, W.; Li, X.; Guan, C.; Hu, Z.; Zhao, Y.; Li, W.; Jiang, X. LncRNA PCAT6 promotes the proliferation, migration and invasion of pancreatic ductal adenocarcinoma via regulating miR-185-5p/CBX2 axis. Pathol. Res. Pract. 2020, 216, 153074. [Google Scholar] [CrossRef] [PubMed]
  92. Morvaridi, S.; Dhall, D.; Greene, M.I.; Pandol, S.J.; Wang, Q. Role of YAP and TAZ in pancreatic ductal adenocarcinoma and in stellate cells associated with cancer and chronic pancreatitis. Sci. Rep. 2015, 5, 16759. [Google Scholar] [CrossRef] [Green Version]
  93. Xia, D.; Li, X.; Niu, Q.; Liu, X.; Xu, W.; Ma, C.; Gu, H.; Liu, Z.; Shi, L.; Tian, X.; et al. MicroRNA-185 suppresses pancreatic cell proliferation by targeting transcriptional coactivator with PDZ-binding motif in pancreatic cancer. Exp. Ther. Med. 2018, 15, 657–666. [Google Scholar] [CrossRef]
  94. Gao, W.; Wang, J.; Wang, M.; Wang, C. Potential gene and microRNA biomarkers for pancreatic cancer with gene expression analysis. Int. J. Clin. Exp. Pathol. 2016, 9, 1917–1924. [Google Scholar]
  95. Liu, R.; Chen, X.; Du, Y.; Yao, W.; Shen, L.; Wang, C.; Hu, Z.; Zhuang, R.; Ning, G.; Zhang, C.; et al. Serum microRNA expression profile as a biomarker in the diagnosis and prognosis of pancreatic cancer. Clin. Chem. 2012, 58, 610–618. [Google Scholar] [CrossRef] [Green Version]
  96. Al-Shaheri, F.N.; Alhamdani, M.S.S.; Bauer, A.S.; Giese, N.; Büchler, M.W.; Hackert, T.; Hoheisel, J.D. Blood biomarkers for differential diagnosis and early detection of pancreatic cancer. Cancer Treat. Rev. 2021, 96, 102193. [Google Scholar] [CrossRef]
  97. Sawicki, T.; Ruszkowska, M.; Danielewicz, A.; Niedźwiedzka, E.; Arłukowicz, T.; Przybyłowicz, K.E. A review of colorectal cancer in terms of epidemiology, risk factors, development, symptoms and diagnosis. Cancers 2021, 13, 2025. [Google Scholar] [CrossRef]
  98. Zhang, W.; Sun, Z.; Su, L.; Wang, F.; Jiang, Y.; Yu, D.; Zhang, F.; Sun, Z.; Liang, W. miRNA-185 serves as a prognostic factor and suppresses migration and invasion through Wnt1 in colon cancer. Eur. J. Pharmacol. 2018, 825, 75–84. [Google Scholar] [CrossRef]
  99. Dong-Xu, W.; Jia, L.; Su-Juan, Z. MicroRNA-185 is a novel tumor suppressor by negatively modulating the Wnt/β-catenin pathway in human colorectal cancer. Indian J. Cancer 2015, 52, e182–e185. [Google Scholar] [CrossRef]
  100. Liu, C.; Ji, L.; Song, X. Long non coding RNA UCA1 contributes to the autophagy and survival of colorectal cancer cells via sponging miR-185-5p to up-regulate the WISP2/β-catenin pathway. RSC Adv. 2019, 9, 14160–14166. [Google Scholar] [CrossRef] [Green Version]
  101. Liu, M.; Lang, N.; Chen, X.; Tang, Q.; Liu, S.; Huang, J.; Zheng, Y.; Bi, F. MiR-185 targets RhoA and Cdc42 expression and inhibits the proliferation potential of human colorectal cells. Cancer Lett. 2011, 301, 151–160. [Google Scholar] [CrossRef] [PubMed]
  102. Liao, J.M.; Lu, H. Autoregulatory suppression of c-Myc by miR-185-3p. J. Biol. Chem. 2011, 286, 33901–33909. [Google Scholar] [CrossRef] [Green Version]
  103. Zhou, C.; Kong, W.; Ju, T.; Xie, Q.; Zhai, L. MiR-185-3p mimic promotes the chemosensitivity of CRC cells via AQP5. Cancer Biol. Ther. 2020, 21, 790–798. [Google Scholar] [CrossRef] [PubMed]
  104. Zhang, Z.; Liu, X.; Feng, B.; Liu, N.; Wu, Q.; Han, Y.; Nie, Y.; Wu, K.; Shi, Y.; Fan, D. STIM1, a direct target of microRNA-185, promotes tumor metastasis and is associated with poor prognosis in colorectal cancer. Oncogene 2015, 34, 4808–4820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Afshar, S.; Najafi, R.; Sedighi Pashaki, A.; Sharifi, M.; Nikzad, S.; Gholami, M.H.; Khoshghadam, A.; Amini, R.; Karimi, J.; Saidijam, M. MiR-185 enhances radiosensitivity of colorectal cancer cells by targeting IGF1R and IGF2. Biomed. Pharmacother. 2018, 106, 763–769. [Google Scholar] [CrossRef]
  106. Lu, Z.J.; Lu, L.G.; Tao, K.Z.; Chen, D.F.; Xia, Q.; Weng, J.J.; Zhu, F.; Wang, X.P.; Zheng, P. MicroRNA-185 suppresses growth and invasion of colon cancer cells through inhibition of the hypoxiainducible factor-2α pathway in vitro and in vivo. Mol. Med. Rep. 2014, 10, 2401–2408. [Google Scholar] [CrossRef] [Green Version]
  107. Yuan, M.; Zhang, X.; Zhang, J.; Wang, K.; Zhang, Y.; Shang, W.; Zhang, Y.; Cui, J.; Shi, X.; Na, H.; et al. DC-SIGN–LEF1/TCF1–miR-185 feedback loop promotes colorectal cancer invasion and metastasis. Cell Death Differ. 2020, 27, 379–395. [Google Scholar] [CrossRef]
  108. Li, J.; Liu, H.; Zou, L.; Ke, J.; Zhang, Y.; Zhu, Y.; Yang, Y.; Gong, Y.; Tian, J.; Zou, D.; et al. A functional variant in GREM1 confers risk for colorectal cancer by disrupting a hsa-miR-185-3p binding site. Oncotarget 2017, 8, 61318–61326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Liu, X.; Meng, X.; Peng, X.; Yao, Q.; Zhu, F.; Ding, Z.; Sun, H.; Liu, X.; Li, D.; Lu, Y.; et al. Impaired AGO2/miR-185-3p/NRP1 axis promotes colorectal cancer metastasis. Cell Death Dis. 2021, 12, 390. [Google Scholar] [CrossRef]
  110. Vivekanandhan, S.; Mukhopadhyay, D. Genetic status of KRAS influences transforming growth factor-beta (TGF-β) signaling: An insight into neuropilin-1 (NRP1) mediated tumorigenesis. Semin. Cancer Biol. 2019, 54, 72–79. [Google Scholar] [CrossRef]
  111. Tang, H.I.; Deng, M.; Liao, Q.J. Expression and clinical significance of mir-185 in tissues of colon carcinoma. J. Med. Sci. Cent. S. China 2011, 5, 1. [Google Scholar]
  112. Zhang, N.; Hu, X.; Du, Y.; Du, J. The role of miRNAs in colorectal cancer progression and chemoradiotherapy. Biomed. Pharmacother. 2021, 134, 111099. [Google Scholar] [CrossRef]
  113. Bobowicz, M.; Skrzypski, M.; Czapiewski, P.; Marczyk, M.; Maciejewska, A.; Jankowski, M.; Szulgo-Paczkowska, A.; Zegarski, W.; Pawłowski, R.; Polańska, J.; et al. Prognostic value of 5-microRNA based signature in T2-T3N0 colon cancer. Clin. Exp. Metastasis 2016, 33, 765–773. [Google Scholar] [CrossRef] [Green Version]
  114. Matsumura, T.; Sugimachi, K.; Iinuma, H.; Takahashi, Y.; Kurashige, J.; Sawada, G.; Ueda, M.; Uchi, R.; Ueo, H.; Takano, Y.; et al. Exosomal microRNA in serum is a novel biomarker of recurrence in human colorectal cancer. Br. J. Cancer 2015, 113, 275–281. [Google Scholar] [CrossRef] [PubMed]
  115. Tan, K.; Leong, S.M.; Kee, Z.; Caramat, P.V.; Teo, J.; Blanco, M.; Koay, E.; Cheong, W.K.; Soh, T.I.; Yong, W.P.; et al. Longitudinal monitoring reveals dynamic changes in circulating tumor cells (CTCs) and CTC-associated miRNAs in response to chemotherapy in metastatic colorectal cancer patients. Cancer Lett. 2018, 423, 1–8. [Google Scholar] [CrossRef]
  116. Sur, D.; Balacescu, L.; Cainap, S.S.; Visan, S.; Pop, L.; Burz, C.; Havasi, A.; Buiga, R.; Cainap, C.; Irimie, A.; et al. Predictive efficacy of MiR-125b-5p, MiR-17-5p, and MiR-185-5p in liver metastasis and chemotherapy response among advanced stage colorectal cancer patients. Front. Oncol. 2021, 11, 651380. [Google Scholar] [CrossRef]
  117. Maciel-Dominguez, A.; Swan, D.; Ford, D.; Hesketh, J. Selenium alters miRNA profile in an intestinal cell line: Evidence that miR-185 regulates expression of GPX2 and SEPSH2. Mol. Nutr. Food Res. 2013, 57, 2195–2205. [Google Scholar] [CrossRef]
  118. Akçakaya, P.; Ekelund, S.; Kolosenko, I.; Caramuta, S.; Ozata, D.M.; Xie, H.; Lindforss, U.; Olivecrona, H.; Lui, W.O. miR-185 and miR-133b deregulation is associated with overall survival and metastasis in colorectal cancer. Int. J. Oncol. 2011, 39, 311–318. [Google Scholar] [PubMed]
  119. Baldi, S.; Khamgan, H.; Qian, Y.; Wu, H.; Zhang, Z.; Zhang, M.; Gao, Y.; Safi, M.; Al-Radhi, M.; Zuo, Y.F. Downregulated ARID1A by miR-185 is associated with poor prognosis and adverse outcomes in colon adenocarcinoma. Front. Oncol. 2021, 11, 679334. [Google Scholar] [CrossRef] [PubMed]
  120. Zhang, H.; Li, W.; Nan, F.; Ren, F.; Wang, H.; Xu, Y.; Zhang, F. MicroRNA expression profile of colon cancer stem-like cells in HT29 adenocarcinoma cell line. Biochem. Biophys. Res. Commun. 2011, 404, 273–278. [Google Scholar] [CrossRef]
  121. Zhang, B.; Wang, X.; Wang, Y. Altered gene expression and miRNA expression associated with cancerous IEC-6 cell transformed by MNNG. J. Exp. Clin. Cancer Res. 2009, 28, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Cao, C.; Zhang, J.; Yang, C.; Xiang, L.; Liu, W. Silencing of long noncoding RNA UCA1 inhibits colon cancer invasion, migration and epithelial-mesenchymal transition and tumour formation by upregulating miR-185-5p in vitro and in vivo. Cell Biochem. Funct. 2020, 38, 176–184. [Google Scholar] [CrossRef]
  123. Wang, M.; Zheng, Q.; Zhao, Z.; Deng, H.; Zhang, Q.; Yao, H. HES5 activates long noncoding RNA UCA1 to induce colorectal cancer progression by modulating miR-185/NOTCH3 signaling. Gastroenterol. Res. Pract. 2021, 2021, 7249818. [Google Scholar] [CrossRef]
  124. Ou, C.; Sun, Z.; He, X.; Li, X.; Fan, S.; Zheng, X.; Peng, Q.; Li, G.; Li, X.; Ma, J. Targeting YAP1/LINC00152/FSCN1 signaling axis prevents the progression of colorectal cancer. Adv. Sci. 2020, 7, 1901380. [Google Scholar] [CrossRef] [Green Version]
  125. Zhuang, S.-T.; Cai, Y.-J.; Liu, H.-P.; Qin, Y.; Wen, J.F. LncRNA NEAT1/miR-185-5p/IGF2 axis regulates the invasion and migration of colon cancer. Mol. Genet. Genom. Med. 2020, 8, e1125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Zhu, Y.; Qiao, L.; Zhou, Y.; Ma, N.; Wang, C.; Zhou, J. Long non-coding RNA FOXD2-AS1 contributes to colorectal cancer proliferation through its interaction with microRNA-185-5p. Cancer Sci. 2018, 109, 2235–2242. [Google Scholar] [CrossRef] [Green Version]
  127. Sun, W.; Nie, W.; Wang, Z.; Zhang, H.; Li, Y.; Fang, X. Lnc haglr promotes colon cancer progression through sponging mir-185-5p and activating cdk4 and cdk6 in vitro and in vivo. Onco Target. Ther. 2020, 13, 5913–5925. [Google Scholar] [CrossRef]
  128. Zhang, L.; Dong, X.; Yan, B.; Yu, W.; Shan, L. CircAGFG1 drives metastasis and stemness in colorectal cancer by modulating YY1/CTNNB1. Cell Death Dis. 2020, 11, 542. [Google Scholar] [CrossRef]
  129. Lu, W.; Huang, Z.; Wang, J.; Liu, H. Long non-coding RNA DANCR accelerates colorectal cancer progression via regulating the miR-185-5p/HMGA2 axis. J. Biochem. 2022, 171, 389–398. [Google Scholar] [CrossRef] [PubMed]
  130. Yu, M.; Shi, C.; Xu, D.; Lin, X.; Ji, T.; Shi, Z.; Zhuge, X.; Zhuo, S.; Yang, Q. LncRNA ASB16-AS1 drives proliferation, migration, and invasion of colorectal cancer cells through regulating miR-185-5p/TEAD1 axis. Cell Cycle 2021, 21, 1–11. [Google Scholar] [CrossRef] [PubMed]
  131. Xiang, Y.; Tian, Q.; Guan, L.; Niu, S. The dual role of miR-186 in cancers: Oncomir battling with tumor suppressor miRNA. Front. Oncol. 2020, 10, 233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Dagogo-Jack, I.; Shaw, A.T. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 2017, 15, 81–94. [Google Scholar] [CrossRef] [PubMed]
  133. Erhard, F.; Haas, J.; Lieber, D.; Malterer, G.; Jaskiewicz, L.; Zavolan, M.; Dölken, L.; Zimmer, R. Widespread context dependency of microRNA-mediated regulation. Genome Res. 2014, 24, 906–919. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of miR-185 maturation. The gene is located on chromosome 22 (GRC genome assembly number: GRCh38.p14; gene ID: 406961). The stem-loop pre-miRNA, the duplex and the mature sequences are shown. Mostly, the red-labeled strand acts as guide. However, this does change in some instances. In consequence, two mature sequences exist. Sequences were downloaded from miRBase (www.mirbase.com; accessed on 21 September 2022) and the NCBI Gene Database (www.ncbi.nlm.nih.gov; accessed on 21 September 2022).
Figure 1. Schematic representation of miR-185 maturation. The gene is located on chromosome 22 (GRC genome assembly number: GRCh38.p14; gene ID: 406961). The stem-loop pre-miRNA, the duplex and the mature sequences are shown. Mostly, the red-labeled strand acts as guide. However, this does change in some instances. In consequence, two mature sequences exist. Sequences were downloaded from miRBase (www.mirbase.com; accessed on 21 September 2022) and the NCBI Gene Database (www.ncbi.nlm.nih.gov; accessed on 21 September 2022).
Ncrna 08 00067 g001
Figure 2. Roles of miR-185 as a tumor suppressor miR or as an oncomiR in digestive tract cancers. Grey boxes show the genes and pathways that are targeted by miR-185. miR-185 has been implicated in the regulation of the tumor-related processes listed on the far left or right. Also, different upstream regulators of miR-185 are presented.
Figure 2. Roles of miR-185 as a tumor suppressor miR or as an oncomiR in digestive tract cancers. Grey boxes show the genes and pathways that are targeted by miR-185. miR-185 has been implicated in the regulation of the tumor-related processes listed on the far left or right. Also, different upstream regulators of miR-185 are presented.
Ncrna 08 00067 g002
Figure 3. LncRNAs and circRNAs are shown which act as major upstream regulators of miR-185 in digestive tract cancers and drive cancer progression by suppressing miR-185.
Figure 3. LncRNAs and circRNAs are shown which act as major upstream regulators of miR-185 in digestive tract cancers and drive cancer progression by suppressing miR-185.
Ncrna 08 00067 g003
Table 1. Target genes of miR-185. Genes or pathways affected by miR-185-5p are labeled in red, the ones targeted by miR-185-3p in blue. Genes or pathways for which this information is not available from the literature are indicated in black letters.
Table 1. Target genes of miR-185. Genes or pathways affected by miR-185-5p are labeled in red, the ones targeted by miR-185-3p in blue. Genes or pathways for which this information is not available from the literature are indicated in black letters.
Cancer TissueIdentified by
In Vitro Experiments
Predicted
from Data
Verified by
In Vivo Experiments
Mouth and pharynxALOX12B, AQP3,CASP-14,CCND2, FOXD3, HOXC6, RAB14,SMAD7, TGF-β1/mTOR axis, WNT2B, YWHAZ, ZNF703, Akt/CASP-9 pathway, Akt/NF-κB pathway, Wnt2b/β-catenin/c-Myc pathway CCND2, FOXD3, HOXC6, YWHAZ, ZNF703, Akt/CASP-9 pathway, Akt/NF-κB pathway
EsophagusKLF3, KLK5,RAGE, SIX1 KLK5,RAGE
StomachARC, BCL-2, CASP-3, CASP-8, CCND2, CTSD,DNMT1, EZH2,GST-π, MDR1/P-gp, MRP-1,RHOA, TGF-β1, TRIM29, XIAP, Wnt/β-catenin pathwayAQP5, ESRRA, RAC3, RGS14, ZNFN1A4ARC,CTSD, DNMT1, EZH2, RHOA
LiverAQP11, AKT, CDC42, CDH1, DNMT1, DNMT3A, DNMT3B, ELK1, KIF14,KNL1, MEG3,MKI67, PLAC8,ROCK2, SIX2, SPC25, VIM, Akt1 signaling pathway, DNMT1/PTEN/Akt pathway, ITGB5/β-catenin- and PLAC8/Wnt/β-catenin pathway CDC42, DNMT1/PTEN/Akt pathway, ITGB5/β-catenin pathway
PancreasCBX2, CCND2, TAFAZZINCORO2B, NTRK3CBX2, CCND2,
TAFAZZIN
Colon or
rectum
AQP5, ARID1A,c-MYC, CDC42, CDK4, CDK6, DC-SIGN, FSCN1, GREM1, HIF-2α, HMGA2, IGF1R, IGF2, MAPK14, NOTCH3, NRP1, RHOA, STIM1, TEAD1, YY1, MAPKAPK2/HSP27 pathway, WISP2/β-catenin pathway, Wnt/β-catenin pathway AQP5, CDC42, CDK4, CDK6, DC-SIGN, FSCN1, HIF-2α, HMGA2, IGF2, MAPK14, NRP1, STIM1, TEAD1,YY1, MAPKAPK2/HSP27 pathway, WISP2/β-catenin pathway
Table 2. Example for the fragmented description of miR-185 regulation and activity in oral cancer.
Table 2. Example for the fragmented description of miR-185 regulation and activity in oral cancer.
Upstream
Regulator
ProcessAffected Gene or Pathway/FunctionRef.
FOXD3-AS1Reduction of miR-185 levelFOXD3[26]
irc0058106Wnt2b/β-catenin/c-Myc pathway[27]
LINC00958YWHAZ[28]
PDIA3PCCND2[30]
LSINCT5ZNF703[31]
KCNQ1OT1Increased tumorigenesis and reduced apoptosis[32]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Babaeenezhad, E.; Naghibalhossaini, F.; Rajabibazl, M.; Jangravi, Z.; Hadipour Moradi, F.; Fattahi, M.D.; Hoheisel, J.D.; Sarabi, M.M.; Shahryarhesami, S. The Roles of microRNA miR-185 in Digestive Tract Cancers. Non-Coding RNA 2022, 8, 67. https://doi.org/10.3390/ncrna8050067

AMA Style

Babaeenezhad E, Naghibalhossaini F, Rajabibazl M, Jangravi Z, Hadipour Moradi F, Fattahi MD, Hoheisel JD, Sarabi MM, Shahryarhesami S. The Roles of microRNA miR-185 in Digestive Tract Cancers. Non-Coding RNA. 2022; 8(5):67. https://doi.org/10.3390/ncrna8050067

Chicago/Turabian Style

Babaeenezhad, Esmaeel, Fakhraddin Naghibalhossaini, Masoumeh Rajabibazl, Zohreh Jangravi, Forouzan Hadipour Moradi, Mohammad Davood Fattahi, Jörg D. Hoheisel, Mostafa Moradi Sarabi, and Soroosh Shahryarhesami. 2022. "The Roles of microRNA miR-185 in Digestive Tract Cancers" Non-Coding RNA 8, no. 5: 67. https://doi.org/10.3390/ncrna8050067

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop