Abstract
MicroRNAs (miRNAs) are one abundant class of small, endogenous non-coding RNAs, which regulate various biological processes by inhibiting expression of target genes. miRNAs have important functional roles in carcinogenesis and development of colorectal cancer (CRC), and emerging evidence has indicated the feasibility of miRNAs as robust cancer biomarkers. This review summarizes the progress in miRNA-related research, including study of its oncogene or tumour-suppressor roles and the advantages of miRNA biomarkers for CRC diagnosis, treatment and recurrence prediction. Along with analytical technique improvements in miRNA research, use of the emerging extracellular miRNAs is feasible for CRC diagnosis and prognosis.
Keywords: microRNAs, colorectal cancer, biomarkers, diagnosis and prognosis
1. Introduction
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers and a leading cause of death worldwide [1–3]. One of the important factors predicting survival in CRC patients is metastasis. In contrast to patients with stage II (no lymph node metastases) disease, the 5-year survival for patients with stage III CRC (with the presence of cancer cells in tumour-draining lymph nodes) declines more than 20% [4]. Nowadays, patients are diagnosed with CRC with a trend toward younger age [5]. In the last decade, CRC incidence rates increased by 22% and CRC death rates increased by 13% among adults aged less than 50 years in the USA [6]. However, the precise aetiologic factors of these onset cases have yet to be elucidated. As CRC develops slowly from removable precancerous lesions, early screening can reduce the incidence and mortality of this malignancy, and relevant diagnosis or prognosis biomarkers will be helpful for assessing tumour initiation, progression and response to treatment in CRC [7,8].
MicroRNAs (miRNAs) are small non-coding RNAs with 20–22 nucleotides, which participate in multiple biological processes [9]. miRNAs have been reported to be involved in occurrence and progression of various types of cancers, including brain, lung, breast, liver, prostate and colorectal cancer [10–16]. By targeting the 3′UTR of target genes, miRNAs influence a number of aspects of cancer cells by functioning as tumour suppressors or oncogenes, including mediating cell proliferation and migration [12,15], autophagy [10,14], apoptosis [11], metabolic shift [17], epithelial–mesenchymal transition [13,18] and radiosensitivity [19].
miRNAs have already entered into cancer clinics as promising biomarkers [8]. Dysregulation phenotypes of miRNAs have been associated with CRC [16,20–22]. The candidate miRNA biomarkers are usually screened through exploring differential expression profiling of miRNAs in CRC tissues compared with the paired neighbouring non-cancerous colorectal tissues.
In this review, we summarize the differential expressions of miRNAs and their functions in CRC, and miRNA roles in CRC diagnosis, treatment and recurrence prediction. Moreover, the emerging extracellular miRNAs are illustrated as CRC biomarkers, owing to improvements in the detection approaches for miRNAs.
2. Aberrant miRNA expressions and roles in CRC
Aberrant expressions of miRNAs and their roles in various biological processes have been revealed to be associated with CRC carcinogenesis. miRNAs function as either oncogenes or tumour suppressors by regulating different targets (table 1). For example, miR-18a [25], miR-155 [23,44] and miR-205-5p [18,24] repress proliferation, migration and invasion of CRC cells, whereas miR-494 [38], miR-598 [39] and miR-17-3p [40] promote the abilities of cell proliferation, migration and invasion. miR-106a [41] and miR-7 [26] relate to apoptosis of CRC cells or the resistance to apoptosis. miR-221 and miR-214 reduce autophagy in CRC cells [42,43]. miR-192/215 [27] and miR-19b-1 [28] have regulatory roles in metabolic pathways. Other miRNAs regulating different targets are also included in table 1 [29–37].
Table 1.
miRNA | dysregulation | target gene | effects | ref. |
---|---|---|---|---|
miR-155 | downregulated | CTHRC1 | suppress cell proliferation, promote cell cycle arrest and apoptosis | [23] |
miR-205-5p | downregulated | ZEB1 | inhibit epithelial to mesenchymal transition | [18,24] |
miR-18a | downregulated | CDC42 | inhibit colorectal cancer cell growth and death | [25] |
miR-7 | downregulated | YY1 | resensitization to fas/FasL-apoptosis | [26] |
miR-192/215 | upregulated | SRPX2 | facilitates cell glycolysis | [27] |
miR-19b-1 | downregulated | ACSL/SCD | inhibit invasion in colon cancer cells | [28] |
miR-30a | downregulated | metadherin | inhibit cell migration and invasion | [29] |
miR-744 | downregulated | Notch1 | inhibit cell proliferation and invasion | [30] |
miR-383 | downregulated | PAX6 | inhibit cell proliferation and invasion | [31] |
miR-1271 | downregulated | Capn4 | suppress cell proliferation and invasion | [32] |
miR-186-5p | downregulated | ZEB1 | inhibit cell proliferation, metastasis and epithelial to mesenchymal transition | [33] |
miR-511 | downregulated | HDGF | reduce cell proliferation and invasion | [34] |
miR-374b | downregulated | LRH-1 | inhibit cell proliferation and invasion | [35] |
miR-216a-3p | downregulated | COX-2 and ALOX5 | suppress cell proliferation | [36] |
miR-1273g-3p | upregulated | CNR1 | promote cell proliferation, migration and invasion | [37] |
miR-494 | upregulated | APC | promote cell growth | [38] |
miR-598 | upregulated | INPP5E | promote cell proliferation and cell cycle progression | [39] |
miR-17-3p | upregulated | Par4 | promote cell proliferation and reduce apoptosis | [40] |
miR-106a | upregulated | PTEN | promote cell proliferation and reduce apoptosis | [41] |
miR-221 | upregulated | TP53INP1 | promote cell proliferation and reduce apoptosis | [42] |
miR-214 | downregulated | ATG12 | promote radiosensitivity by inhibiting IR-induced autophagy | [43] |
miR-26a | upregulated | PDHX | inhibit glucose metabolism | [16] |
miR-508 induces the stem-like/mesenchymal subtype in CRC by affecting the expression of cadherin CDH1 and the transcription factors ZEB1, SALL4 and BMI1 [45]. miR-15A and 16-1 reduce the chemotaxis of IgA(+) B cells and activate signalling pathways required for B-cell-mediated immune suppression in CRC [46]. miR-21-5p exhibits epigenetic effects in CRC by blocking the activation of DNA demethylation [47].
Owing to the high-throughput genome-wide profiling and comprehensive screening technologies, novel miRNA signatures [48,49] and multiple miRNA–mRNA regulatory networks [50,51] have been discovered in CRC. miRNA-mediated genes and signalling pathways are also explored for their associations with CRC. NF-κB regulates immune response and inflammation processes, and is associated with multiple miRNAs such as miR-150-5p, miR-195-5p and miR-203a in carcinogenesis [52]. These studies indicate miRNAs and their targets of action are tightly involved in CRC progression, which gives a promising outlook for the identification of miRNA biomarkers for CRC.
3. miRNAs for CRC diagnosis, treatment and recurrence prediction
miRNAs have shown great clinical value in the diagnosis, treatment and prognosis of CRC (figure 1). Developing appropriate miRNA biomarkers is essential for early stage CRC diagnosis. The differential miRNAs and miRNA-regulated genes were screened in early stage CRC tissues, precancerous lesions and colonic intraepithelial neoplasia by RNA sequencing [53]. miR-548c-5p, miR-548i and miR-548am-5p were identified as the top three of 26 differentially expressed miRNAs with regard to lymph node metastasis [53]. But some other studies report no significant correlations of miRNA-expression patterns with CRC tumour stage by a miRNA microarray analysis [54]. The difference in conclusions is probably due to different detection approaches and tumour sample heterogeneity.
Through bioinformatics analysis from the Cancer Genome Atlas, five significantly changed miRNAs (miR-32, miR-181b, miR-193b, miR-195 and miR-411) were uncovered in T1 and T2 CRCs with versus without lymph node metastases [55]. This five-miRNA signature was validated with a greater degree of accuracy in two cohorts of patients with T1 cancers and untreated patients [55]. In addition, miRNAs are correlated with molecular histological markers (such as Ki-67 and CD34), which is helpful to determine cell proliferation and angiogenesis in CRC development [56]. New efforts have been made to identify differential expression patterns of miRNAs in genotype-specific CRC. Significantly higher levels of miR-31 and obvious lower miR-373 is present in BRAF-mutated tumours compared with wild-type tumours [57]. Regrettably, there was no difference in expression levels between KRAS- and BRAF-mutated tumours in this study. However, it was a good attempt to investigate miRNA signatures for clinico-pathological differences between KRAS- and BRAF-mutated CRCs.
Furthermore, miRNAs have great potential in CRC treatment and could help to overcome the resistance to cancer therapy. For instance, miR-214 enhances CRC radiosensitivity by inhibiting autophagy in CRC cells [43]. The overexpression of miR-143 is related to the oxidative stress and cell death in CRC cells, which might circumvent resistance of CRC cells to oxaliplatin [58]. miR-195 is able to desensitize CRC cells to 5-fluorouracil (5-FU) [59,60]. Meanwhile, a miR-129 mimic is reported to enhance efficacy to eliminate resistance to 5-FU in CRC stem cells [61]. It is notable that the miR-129 mimic can be delivered to cancer cells without any transfection reagents, including lipids, viral vectors and nanoparticles. Another trial used miR-20a-loading nanoparticles to target liver sinusoidal endothelial cells, and showed a great reduction of liver metastasis of CRC both in vitro and in vivo [62]. All the trials indicate promising miRNA-based therapy for CRC. Moreover, small molecules such as polyamine derivatives have also been developed as miRNA interfering agents to tackle the overexpression of oncogenic miRNAs [63].
So far, miRNAs are promising as biomarkers with prognostic and predictive values [64,65]. A recent study has confirmed an eight-miRNA signature is feasible for predicting tumour recurrence of CRC patients in stages II and III by using three independent genome-wide miRNA-expression profiling datasets [48]. In this study, miRNA biomarkers for CRC were screened from multiple clinical cohorts totalling 736 patients, including patients from the publicly available dataset and two clinical validation cohorts of CRC patients who underwent surgery without neoadjuvant chemotherapy. From 25 miRNAs identified, eight candidates were selected with top statistical significance (p-value < 0.2), including hsa-mir-191, hsa-mir-200b, hsa-mir-30b, hsa-mir-30c2, hsa-mir-33a, hsa-mir-362, hsa-mir-429 and hsa-mir-744, which were further validated in fresh frozen tissues [48]. Moreover, miRNAs from tumour-adjacent mucosa have been studied for the prediction of relapse risk after curative treatment. Four miRNAs (miR-18a, miR-21, miR-182 and miR-183) were found with coordinate deregulation in the normal mucosa adjacent to the tumour, which is predictive of relapse within 55 months from curative surgery in 48 localized CRC patients who underwent radical tumour resection [66].
4. Emerging extracellular miRNAs used for CRC biomarkers
miRNAs can be detected not only in tissue samples but also in extracellular samples including faeces and blood (table 2). miRNAs from different blood or faeces samples were useful for CRC diagnosis or prognosis [67–70,72–75,81]. miRNAs have been reported to be relatively stable in a variety of biological fluids and even formalin-fixed paraffin-embedded tissues [82,83]. In view of this, miRNAs present as promising targets for non-invasive biomarker development in CRC and are attractive for eventual clinical translation. miRNAs such as miR-129 with a high expression level in CRC plasma [84] and miR-24-2 with a low level in CRC serum [71] can be potential positive or negative biomarkers in the diagnosis of CRC patients.
Table 2.
source | miRNA | case (n) | control (n) | sensitivity (%) | specificity (%) | AUC (95% CI) | detection method | clinical use | ref. |
---|---|---|---|---|---|---|---|---|---|
plasma | miR-21 | 31 | 34 | 65 | 85 | — | qPCR | diagnosis | [67] |
186 | 53 | 82.8 | 90.6 | 0.919 (0.87–0.96) |
qPCR | diagnosis and prognosis | [68] | ||
miR-6826 | 93 | — | — | — | 3.670 | qPCR | prediction for a poor response to vaccine treatment | [69] | |
miR-122 | 543 | — | — | — | — | array microRNA cards | prognosis | [70] | |
serum | miR-24-2 | 228 | 68 | — | — | — | qPCR | diagnosis | [71] |
miR-139-3p | 117 | 90 | 96.6 | 97.8 | 0.9935 | qPCR | diagnosis | [72] | |
miR-139-5p | 53 | — | 64 | 80 | 0.59–0.87 | qPCR | prognosis | [73] | |
miR-135a-5p | 60 | 40 | — | — | 0.832 (0.73–0.93) |
qPCR | diagnosis | [74] | |
60 | 50 | 0.875 (0.80–0.95) |
|||||||
miR-203 | 330 | — | — | — | — | qPCR | prognosis and metastasis prediction | [75] | |
(miR-21 + miR-29 + miR-92 +miR-125 +miR-223) |
85 | 78 | 84.7 | 98.7 | 0.952 | qPCR | diagnosis | [76] | |
miR-24 | 111 | 130 | 78.38 | 83.85 | 0.839 | qPCR | early detection | [77] | |
miR-320a | 92.79 | 73.08 | 0.886 | ||||||
miR-423-5p | 91.89 | 70.77 | 0.833 | ||||||
(miR-24 + miR-320a +miR-423-5p) |
92.79 | 70.77 | 0.899 | ||||||
plasma exosomes | miR-21 | 326 | — | — | — | — | TaqMan miRNA assays | prediction of recurrence and poor prognosis in CRC patients with TNM stage II, III or IV. | [78] |
miR-6803-5p | 168 | — | — | — | — | qPCR | diagnosis and prognosis | [79] | |
miR-17-5p | 18 | 10 | — | — | 0.897 (0.80–0.99) |
qPCR | prognosis | [80] | |
11 | 10 | — | — | 0.841 (0.72–0.96) |
|||||
miR-92a-3p | 18 | 10 | — | — | 0.845 (0.72–0.97) |
||||
11 | 10 | — | — | 0.854 (0.74–0.97) |
|||||
saliva | miR-21 | 34 | 34 | 97 | 91 | — | qPCR | diagnosis | [67] |
faecal | miR-4478 | 40 | 16 | — | — | — | SYBR Green miScript PCR system | diagnosis | [81] |
miR-1295b-3p | 40 | 16 | — | — | — |
More studies were performed to profile novel circulating miRNA biomarkers for CRC. A pilot study identified five miRNAs, including miR-31, miR-141, miR-224-3p, miR-576-5p and miR-4669, to be significantly different in sera between patients with colon cancer and healthy controls, indicating a miRNA panel for diagnosis of CRC [85]. Faecal sampling is also an ideal non-invasive alternative for detection [86]. Several miRNAs, including miR-29a [87], miR-223 [88], miR-224, miR-106a [89] and miR-135b [90], are detectable in the faeces and could be informative biomarkers for screening and diagnosis of CRC. A faecal miRNA test combined with routine immunochemical faecal occult blood test (iFOBT) is helpful to identify CRC patients from those with negative iFOBT results and may improve the sensitivity to detect CRC [89].
More and more studies focus on circulating exosomal miRNAs [91–97]. Exosomes represent a type of extracellular vesicle formed from endosomal membrane with encapsulated cystolic contents, containing proteins, mRNAs and miRNAs [98,99]. miRNA-containing exosomes have been isolated from various body fluids, and through the exosomal pathway, miRNAs can be sampled from donor cells and transferred between cells [100]. Circulating exosomal miRNAs are considered as novel diagnostic and prognostic biomarkers for CRC [101,102]. For example, levels of miR-17-5p and miR-92a-3p isolated from serum exosomes were identified to associate with pathologic stages and grades of the CRC patients [80]. High levels of serum exosomal miR-6803-5p were observed in CRC patients, which are correlated with TNM stage (a staging system to describe the amount and spread of tumour in a patient's body, in which T describes the size of the original tumour and whether it has invaded nearby tissue, N describes nearby lymph nodes that are involved and M describes metastasis), lymph node metastasis, liver metastasis as well as poor overall survival and disease-free survival [79]. Serum exosomal miR-4772-3p is considered to be a prognostic biomarker for tumour recurrence in stages II and III CRC patients [103]. Also, miR-21 from serum exosomes is a useful biomarker for the prediction of CRC recurrence and poor prognosis with each tumour stage including TNM stages II, III or IV [78].
5. Advantages and disadvantages of miRNA biomarkers
Despite the great success in decreasing the incidence and mortality of CRC that colonoscopy has achieved [104,105], the process of examination is not friendly enough, being accompanied by discomfort, fear of pain and embarrassment [106,107]. At present, the commonly used plasma biomarkers in CRC detection are carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9), but the practical application of both is limited due to their low sensitivity and specificity [108,109]. Emerging evidence indicates that miRNA is a potential choice for CRC detection compared with traditional blood-based biomarkers such as CEA and CA19-9.
A publication evaluated the diagnostic efficiency of miRNAs and traditional biomarkers such as CEA and CA19-9 [76]. A total of 9936 CRC patients and 7935 healthy controls were included in this study, and the results showed that the concentrations of CEA and CA19-9 were both elevated in the CRC group and significantly different from the healthy control group (p < 0.001 and p = 0.004, respectively), indicating that CEA and CA19-9 were capable for CRC detection. However, the combination of five miRNAs (miR-21 + miR-29 + miR-92 + miR-125 + miR-223) showed a better sensitivity (84.7%) and specificity (98.7%) compared with CEA (69.4% sensitivity, 78.2% specificity) and CA19-9 (65.9% sensitivity, 67.1% specificity) by assessing the sensitivity, specificity, Youden index and the area under the curve (AUC) of the receiver operating curve through meta-analysis methods.
Several miRNAs are potential biomarkers for CRC detection. Through data analysis from 223 CRC patients and 130 healthy controls [77], the sensitivity of miR-24, miR-320a and miR-423-5p for early stage CRC were 77.78%, 90.74% and 88.89%, which were better than CEA and CA19-9 (40.54% and 36.04%, respectively). Moreover, the diagnostic efficiencies of miR-24, miR-320a and miR-423-5p were much higher than traditional CEA and CA19-9 (81.33%, 82.16%, 80.50% versus 70.12%, 66.80%). In addition, the combination of a few miRNAs has been investigated for potential detection of early stage CRC, including a five-serum miRNA detection panel (miR-1246, miR-202-3p, miR-21-3p, miR-1229-3p and miR-532-3p) [110] and a three-plasma miRNA detection panel [111]. In conclusion, miRNAs have been shown to be a powerful tool for CRC detection in a non-invasive manner and have better performance in CRC detection compared with conventional blood-based biomarkers such as CEA and CA19-9.
Even so, miRNA biomarkers still have a number of practical problems that need to be resolved. One of them is that establishment of a sensitive detection method suitable for blood samples is required. The cost is also a concern. On average US$23 is required for a four-miRNAs panel detection in China, while the detection of two known cancer biomarkers, CEA and CA19-9, respectively, costs US$4.6 and US$8.0 for each sample [76].
Moreover, no single miRNA alone has been identified as an ideal CRC biomarker upto now. Similarly to other gene or protein cancer markers, some predictive miRNAs are usually not specific for one kind of cancer. For example, miR-18a is reported to be a tumour suppressor by suppressing CDC42 in CRC [25]. However, miR-18a is also a candidate biomarker for breast cancer and lung cancer, having a significantly higher expression in benign breast biopsy than normal controls [112] and correlating with poor prognosis in patients with non-small cell lung cancer [113]. Similarly, miR-155 inhibits colorectal cancer progression and metastasis [23], while it is significantly overexpressed in breast cancer and cervical cancer with potential as a biomarker [114,115]. Fortunately, a panel of miRNAs can be used to distinguish CRC patients from healthy controls with a relatively high sensitivity and specificity, from testing in a large population of subjects [116]. Therefore, several miRNA combinations are feasible to monitor cancer profiling.
6. Regulation and detection of miRNAs
miRNAs represent a type of approximately 22-nucleotide non-coding RNA molecule. The biogenesis and maturation of miRNAs comprise several regulated steps [117,118]. For the mechanism of aberrant miRNA-expression levels, miRNA processing was taken into consideration, involving various regulatory proteins (such as Drosha [119] and Dicer [120]) and cellular location [121]. In addition, genetic alterations, single nucleotide polymorphisms [122] and epigenetic modification (such as DNA methylation [123,124] and N6-methyl-adenosine [125]) can also affect the processing efficiency of miRNAs. In terms of the extracellular miRNAs, they are subsequently packed in apoptotic bodies, microvesicles and exosomes or bound to RNA-binding proteins, and then released from donor cells [126–128]. From this aspect, factors involved in the secretion processes can affect the abundance of extracellular miRNAs, including triggering secretion of exosomes [129], exosome biogenesis [130] and membrane trafficking [131].
The current gold standard for detecting precancerous adenomas and colorectal cancers is still colonoscopy, but its invasiveness puts patients in great pain. Therefore, miRNA as a non-invasive option has attracted a lot of attention. If miRNAs are to be a reliable and convenient biomarker for the detection and prognosis of CRC, detection methods with high sensitivity and specificity, but also practicality, operability and low price, are indispensable. Conventional detection methods for miRNAs are qPCR, microarray and next-generation sequencing, but no one method is completely ideal for clinical application. In order to meet clinical requirements, various methods such as isothermal amplification techniques and near-infrared technology have been established and have been summarized elsewhere [132].
Here, we focus on several newly reported methods. A method based on ionic liquid (IL; 1-butyl-3-methylimidazolium hexafluorophosphate)-modified chemically activated pencil graphite electrodes (IL/CA/PGEs) [133] has been developed for miR-34a detection, with the advantages of being easy-to-handle, cost-effective, fast and portable. In this platform, the detection limits of miRNA achieved were 109 nM in PBS buffer and 117 nM in diluted FBS medium. The method also shows a good selectivity against other miRNAs. However, its performance with real whole blood has not been characterized, because whole blood detection is an arduous task owing to the existence of interfering factors.
Another energy-transfer-based photoelectrochemical method was developed to detect miR-141 with an integration of entropy-driven toehold-mediated DNA strand displacement (ETSD) reaction with magnetic beads (MBs) [134]. This system improved the sensitivity significantly compared to the absorption and photoluminescence method by elevating the detection limit to 0.5 fM. This protocol was also performed using real blood samples and the average recovery of miR-141 increased from 96 to 108%, which is an acceptable accuracy and reproducibility. Two other published methods, an enzyme-free miRNA target-triggered strand displacement reaction (SDR) amplification strategy-based biosensor [135] and an inductively coupled plasma mass spectrometry (ICP-MS)-based strategy [136], were used for detecting miR-21. In the SDR amplification strategy-based biosensor method, miR-21 was coupled with the redox signal of ferrocene and the detection limit was decreased to 0.34 fM with a good selectivity and reproductivity. Even in real serum samples, it worked well with a recovery value ranging from 92 to 113% [135]. The ICP-MS-based strategy was also a miRNA-triggered system through detection of the isotope 89Y. Target miRNA triggers a chain reaction for alternating hybridization between DNA H1 (bond on UCNPs@DNA probe) and DNA H2, leading to accumulation of ultra-small lanthanide upconversion nanoparticles (UCNPs), which is correlated with the concentration of miR-21. This method provided an alternative option for detecting miR-21 in serum with a detection limit of 41aM [136].
Unlike the two methods above, the third one is a way of direct detection of the target using a tool named molecular beacons [137]. It therefore possesses the advantages of being direct, simple and rapid. However, the sensitivity of the molecular beacons based method is worse owing to the lack of signal amplification. An amplification-free electrochemical method shows good detection sensitivity for exosomal miR-21 from serum samples [138]. The target miRNAs are selectively enriched and isolated through MBs, which are pre-functionalized with capture probes to directly adsorb the targets onto a gold electrode surface. The adsorbed miRNAs are measured electrochemically in the presence of an (Fe(CN)6)4−/3− redox system [138].
In order to improve the quantification of extracellular miRNAs, an optimized protocol has been reported to assess a panel of up to 20 miRNAs in serum samples by using multiplexed Taqman miRNA stem-loop primers in the reverse transcription step [139]. Extra isolation steps for pure exosomes are requisite for detection of exosomal miRNAs, including solely or jointly using ultracentrifugation, size-based isolation, immune-affinity capture, microfluidics-based platforms and water excluding polymer-based methods, depending on structural features of the exosomes [140]. To simplify the procedure of miRNA detection, a detection method without miRNA purification or labelling was provided by Fujii et al. [141]. Collecting extracellular vesicles is not required in this system. Prior to loading a sample, heating is performed at 98°C for 2 min to collapse the extracellular vesicle for the release of miRNA, no matter what kind of sample, blood or urine. Despite its convenience, the downside of this system is insufficient sensitivity, which requires improvement for practical diagnosis. The sensitivity of another purification-free method is, however, better; this is a fishhook probe-based rolling circle amplification (FP-RCA) assay [142]. The linear relationship of FP-RCA showed a good correlation at miRNA concentrations from 100 fM to 10 pM. In addition, the FP-RCA assay is of simplicity, low cost and portability.
7. Perspective
So far, miRNAs have shown a strong potential for their utilization as oncological biomarkers in CRC. Direct non-invasive detection of circulating miRNAs would provide information for diagnosis, prognosis and predictive treatment responses for CRC patients. However, the feasibility of exosomal miRNAs as non-invasive biomarkers in CRC still remains a concern, because of multiple tissue sources of a circulating miRNA. Moreover various confounding socioeconomic, environmental and lifestyle factors have profound effects on miRNA levels [143,144]. To realize the potential of miRNA biomarkers for use in personalized treatment, these factors deserve further exploration of their influences on the miRNA-expression patterns.
Supplementary Material
Data accessibility
This article has no additional data.
Authors' contributions
All authors participated in the preparation of the manuscript, and read and approved the final manuscript.
Competing interests
The authors declare that they have no competing interests.
Funding
This work was supported by the National 863 High Tech Foundation (2014AA020608), the National Key Basic Research Program of China (2013CB911303 and 2011CB910703), the National Natural Science Foundation of China (31470810), the Science & Technology Department of Sichuan Province (2017JY0232), the Health and Family Planning Commission of Sichuan Province (17ZD045) and Chengdu Science and Technology Program (2017-GH02-00062-HZ).
References
- 1.Siegel RL, Miller KD, Jemal A. 2018. Cancer statistics, 2018. CA Cancer J. Clin. 68, 7–30. ( 10.3322/caac.21442) [DOI] [PubMed] [Google Scholar]
- 2.Pan R, et al. 2017. Cancer incidence and mortality: a cohort study in China, 2008–2013. Int. J. Cancer. 141, 1315–1323. ( 10.1002/ijc.30825) [DOI] [PubMed] [Google Scholar]
- 3.Yoshino T, et al. 2018. Pan-Asian adapted ESMO consensus guidelines for the management of patients with metastatic colorectal cancer: a JSMO-ESMO initiative endorsed by CSCO, KACO, MOS, SSO and TOS. Ann. Oncol. 29, 44–70. ( 10.1093/annonc/mdx738) [DOI] [PubMed] [Google Scholar]
- 4.Naxerova K, et al. 2017. Origins of lymphatic and distant metastases in human colorectal cancer. Science 357, 55–60. ( 10.1126/science.aai8515) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Siegel RL, Fedewa SA, Anderson WF, Miller KD, Ma J, Rosenberg PS, Jemal A. 2017. Colorectal cancer incidence patterns in the United States, 1974–2013. J. Natl Cancer Inst. 109, djw322 ( 10.1093/jnci/djw322) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Siegel RL, Miller KD, Fedewa SA, Ahnen DJ, Meester RGS, Barzi A, Jemal A. 2017. Colorectal cancer statistics, 2017. CA Cancer J. Clin. 67, 177–193. ( 10.3322/caac.21395) [DOI] [PubMed] [Google Scholar]
- 7.Chung DC. 2018. Genetic testing and early onset colon cancer. Gastroenterology 154, 788–789. ( 10.1053/j.gastro.2018.02.002) [DOI] [PubMed] [Google Scholar]
- 8.Tanaka T, Tanaka M, Tanaka T, Ishigamori R. 2010. Biomarkers for colorectal cancer. Int. J. Mol. Sci. 11, 3209–3225. ( 10.3390/ijms11093209) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chen B, Li H, Zeng X, Yang P, Liu X, Zhao X, Liang S. 2012. Roles of microRNA on cancer cell metabolism. J. Transl. Med. 10, 228 ( 10.1186/1479-5876-10-228) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Yu Y, et al. 2018. MiR-20a-5p suppresses tumor proliferation by targeting autophagy-related gene 7 in neuroblastoma. Cancer Cell Int. 18, 5 ( 10.1186/s12935-017-0499-2) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Fiori ME, Villanova L, Barbini C, De Angelis ML, De Maria R. 2018. miR-663 sustains NSCLC by inhibiting mitochondrial outer membrane permeabilization (MOMP) through PUMA/BBC3 and BTG2. Cell Death Dis. 9, 49 ( 10.1038/s41419-017-0080-x) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Yang ZX, Zhang B, Wei J, Jiang GQ, Wu YL, Leng BJ, Xing CG. 2018. MiR-539 inhibits proliferation and migration of triple-negative breast cancer cells by down-regulating LAMA4 expression. Cancer Cell Int. 18, 16 ( 10.1186/s12935-018-0512-4) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Hu JG, Zhou SH, Zhang H, Qu J, Xiong XW, Hu JO, Liao CG, Yang SE. 2018. MicroRNA-10b regulates epithelial-mesenchymal transition by modulating KLF4/KLF11/Smads in hepatocellular carcinoma. Cancer Cell Int. 18, 10 ( 10.1186/s12935-018-0508-0) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Sohn EJ. 2018. MicroRNA 200c-3p regulates autophagy via upregulation of endoplasmic reticulum stress in PC-3 cells. Cancer Cell Int. 18, 2 ( 10.1186/s12935-017-0500-0) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wei WT, et al. 2017. miR-422a inhibits cell proliferation in colorectal cancer by targeting AKT1 and MAPK1. Cancer Cell Int. 17, 91 ( 10.1186/s12935-017-0461-3) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Chen B, et al. 2014. MicroRNA-26a regulates glucose metabolism by direct targeting PDHX in colorectal cancer cells. BMC Cancer 14, 443 ( 10.1186/1471-2407-14-443) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Santasusagna S, Moreno I, Navarro A, Munoz C, Martinez F, Hernandez R, Castellano JJ, Monzo M. 2018. miR-328 mediates a metabolic shift in colon cancer cells by targeting SLC2A1/GLUT1. Clin. Transl. Oncol. 20, 1161–1167. ( 10.1007/s12094-018-1836-1) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Gulei D, et al. 2018. The silent healer: miR-205-5p up-regulation inhibits epithelial to mesenchymal transition in colon cancer cells by indirectly up-regulating E-cadherin expression. Cell Death Dis. 9, 66 ( 10.1038/s41419-017-0102-8) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Li TF, Ma J, Han XW, Jia YX, Yuan HF, Shui SF, Guo D. 2018. MicroRNA-320 enhances radiosensitivity of glioma through down-regulation of sirtuin type 1 by directly targeting forkhead box protein M1. Transl. Oncol. 11, 205–212. ( 10.1016/j.tranon.2017.12.008) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Wu WK, Law PT, Lee CW, Cho CH, Fan D, Wu K, Yu J, Sung JJ. 2011. MicroRNA in colorectal cancer: from benchtop to bedside. Carcinogenesis 32, 247–253. ( 10.1093/carcin/bgq243) [DOI] [PubMed] [Google Scholar]
- 21.Dong Y, Wu WK, Wu CW, Sung JJ, Yu J, Ng SS. 2011. MicroRNA dysregulation in colorectal cancer: a clinical perspective. Br. J. Cancer. 104, 893–898. ( 10.1038/bjc.2011.57) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Aslam MI, Taylor K, Pringle JH, Jameson JS. 2009. MicroRNAs are novel biomarkers of colorectal cancer. Br. J. Surg. 96, 702–710. ( 10.1002/bjs.6628) [DOI] [PubMed] [Google Scholar]
- 23.Liu JT, Chen ZY, Xiang JB, Gu XD. 2018. MicroRNA-155 acts as a tumor suppressor in colorectal cancer by targeting CTHRC1 in vitro. Oncol. Lett. 15, 5561–5568. ( 10.3892/ol.2018.8069) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Chen S, Wang Y, Su Y, Zhang L, Zhang M, Li X, Wang J, Zhang X. 2018. miR2055p/PTK7 axis is involved in the proliferation, migration and invasion of colorectal cancer cells. Mol. Med. Rep. 17, 6253–6260. ( 10.3892/mmr.2018.8650) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Humphreys KJ, McKinnon RA, Michael MZ. 2014. miR-18a inhibits CDC42 and plays a tumour suppressor role in colorectal cancer cells. PLoS One 11, e112288 ( 10.1371/journal.pone.0112288) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Pothoulakis C, Torre-Rojas M, Duran-Padilla MA, Gevorkian J, Zoras O, Chrysos E, Chalkiadakis G, Baritaki S. 2018. CRHR2/Ucn2 signaling is a novel regulator of miR-7/YY1/Fas circuitry contributing to reversal of colorectal cancer cell resistance to Fas-mediated apoptosis. Int. J. Cancer 142, 334–346. ( 10.1002/ijc.31064) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zhao J, Xu J, Zhang R. 2018. SRPX2 regulates colon cancer cell metabolism by miR-192/215 via PI3K-Akt. Am. J. Transl. Res. 10, 483–490. [PMC free article] [PubMed] [Google Scholar]
- 28.Cruz-Gil S, et al. 2018. Targeting the lipid metabolic axis ACSL/SCD in colorectal cancer progression by therapeutic miRNAs: miR-19b-1 role. J. Lipid Res. 59, 14–24. ( 10.1194/jlr.M076752) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Jin H, Shi X, Zhao Y, Peng M, Kong Y, Qin D, Lv X. 2018. MicroRNA-30a mediates cell migration and invasion by targeting metadherin in colorectal cancer. Technol. Cancer Res. Treat. 17, 1533033818758108 ( 10.1177/1533033818758108) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Shen J, Li M. 2018. MicroRNA-744 inhibits cellular proliferation and invasion of colorectal cancer by directly targeting oncogene Notch1. Oncol. Res. 26, 1401–1409. ( 10.3727/096504018X15188747585738) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Yan F, Tu Z, Duan L, Wang D, Lin F. 2018. MicroRNA-383 suppresses cell proliferation and invasion in colorectal cancer by directly targeting paired box6. Mol. Med. Rep. 17, 6893–6901. ( 10.3892/mmr.2018.8682) [DOI] [PubMed] [Google Scholar]
- 32.Li J, Xu J, Yan X, Jin K, Li W, Zhang R. 2018. Suppression of Capn4 by microRNA-1271 impedes the proliferation and invasion of colorectal cancer cells. Biomed. Pharmacother. 99, 162–168. ( 10.1016/j.biopha.2017.12.107) [DOI] [PubMed] [Google Scholar]
- 33.Li J, Xia L, Zhou Z, Zuo Z, Xu C, Song H, Cai J. 2018. MiR-186-5p upregulation inhibits proliferation, metastasis and epithelial-to-mesenchymal transition of colorectal cancer cell by targeting ZEB1. Arch. Biochem. Biophys. 640, 53–60. ( 10.1016/j.abb.2018.01.002) [DOI] [PubMed] [Google Scholar]
- 34.He S, Wang G, Ni J, Zhuang J, Zhuang S, Wang G, Ye Y, Xia W. 2018. MicroRNA-511 inhibits cellular proliferation and invasion in colorectal cancer by directly targeting hepatoma-derived growth factor. Oncol. Res. 26, 1355–1363. ( 10.3727/096504018X15154094331876) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Qu R, Hao S, Jin X, Shi G, Yu Q, Tong X, Guo D. 2018. MicroRNA-374b reduces the proliferation and invasion of colon cancer cells by regulation of LRH-1/Wnt signaling. Gene 642, 354–361. ( 10.1016/j.gene.2017.11.019) [DOI] [PubMed] [Google Scholar]
- 36.Wang D, Li Y, Zhang C, Li X, Yu J. 2018. MiR-216a-3p inhibits colorectal cancer cell proliferation through direct targeting COX-2 and ALOX5. J. Cell. Biochem. 119, 1755–1766. ( 10.1002/jcb.26336) [DOI] [PubMed] [Google Scholar]
- 37.Li M, Qian X, Zhu M, Li A, Fang M, Zhu Y, Zhang J. 2018. miR1273 g-3p promotes proliferation, migration and invasion of LoVo cells via cannabinoid receptor 1 through activation of ERBB4/PIK3R3/mTOR/S6K2 signaling pathway. Mol. Med. Rep. 17, 4619–4626. ( 10.3892/mmr.2018.8397) [DOI] [PubMed] [Google Scholar]
- 38.Zhang Y, Guo L, Li YH, Feng GH, Teng F, Li W, Zhou Q. 2018. MicroRNA-494 promotes cancer progression and targets adenomatous polyposis coli in colorectal cancer. Mol. Cancer. 17, 1 ( 10.1186/s12943-017-0753-1) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Li KP, Fang YP, Liao JQ, Duan JD, Feng LG, Luo XZ, Liang ZJ. 2018. Upregulation of miR598 promotes cell proliferation and cell cycle progression in human colorectal carcinoma by suppressing INPP5E expression. Mol. Med. Rep. 17, 2991–2997. ( 10.3892/mmr.2017.8207) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Lu D, Tang L, Zhuang Y, Zhao P. 2018. miR-17-3P regulates the proliferation and survival of colon cancer cells by targeting Par4. Mol. Med. Rep. 17, 618–623. ( 10.3892/mmr.2017.7863) [DOI] [PubMed] [Google Scholar]
- 41.Qin Y, Huo ZB, Song X, Chen X, Tian XP, Wang XJ. 2018. mir-106a regulates cell proliferation and apoptosis of colon cancer cells through targeting the PTEN/PI3 K/AKT signaling pathway. Oncol. Lett. 15, 3197–3201. ( 10.3892/ol.2017.7715) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Liao D, Li T, Ye CG, Zeng LY, Li HH, Pu XX, Ding CC, He ZW, Huang GL. 2018. miR-221 inhibits autophagy and targets TP53INP1 in colorectal cancer cells. Exp. Ther. Med. 15, 1712–1717. ( 10.3892/etm.2017.5522) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hu JL, et al. 2018. Inhibition of ATG12-mediated autophagy by miR-214 enhances radiosensitivity in colorectal cancer. Oncogenesis 7, 16 ( 10.1038/s41389-018-0028-8) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Liu N, Jiang F, Han XY, Li M, Chen WJ, Liu QC, Liao CX, Lv YF. 2018. MiRNA-155 promotes the invasion of colorectal cancer SW-480 cells through regulating the Wnt/beta-catenin. Eur. Rev. Med. Pharmacol. Sci. 22, 101–109. ( 10.26355/eurrev_201801_14106) [DOI] [PubMed] [Google Scholar]
- 45.Yan TT, et al. 2018. miR-508 defines the stem-like/mesenchymal subtype in colorectal cancer. Cancer Res. 78, 1751–1765. ( 10.1158/0008-5472.Can-17-2101) [DOI] [PubMed] [Google Scholar]
- 46.Liu RH, et al. 2018. MicroRNAs 15A and 16-1 activate signaling pathways that mediate chemotaxis of immune regulatory B cells to colorectal tumors. Gastroenterology 154, 637–651. ( 10.1053/j.gastro.2017.09.045) [DOI] [PubMed] [Google Scholar]
- 47.Cheng YW, Chou CJ, Yang PM. 2018. Ten-eleven translocation 1 (TET1) gene is a potential target of miR-21-5p in human colorectal cancer. Surg. Oncol. 27, 76–81. ( 10.1016/j.suronc.2017.12.004) [DOI] [PubMed] [Google Scholar]
- 48.Kandimalla R, et al. 2018. Genome-wide discovery and identification of a novel miRNA signature for recurrence prediction in stage II and III colorectal cancer. Clin. Cancer Res. 24, 3867–3877. ( 10.1158/1078-0432.Ccr-17-3236) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Du BB, Wu DW, Yang XF, Wang T, Shi XL, Lv YC, Zhou ZL, Liu Q, Zhang WS. 2018. The expression and significance of microRNA in different stages of colorectal cancer. Medicine 97, e9635 ( 10.1097/MD.0000000000009635) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Vishnubalaji R, Hamam R, Abdulla MH, Mohammed MAV, Kassem M, Al-Obeed O, Aldahmash A, Alajez NM. 2015. Genome-wide mRNA and miRNA expression profiling reveal multiple regulatory networks in colorectal cancer. Cell Death Dis. 6, e1614 ( 10.1038/cddis.2014.556) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Xu P, Wang JH, Sun B, Xiao ZD. 2018. Integrated analysis of miRNA and mRNA expression data identifies multiple miRNAs regulatory networks for the tumorigenesis of colorectal cancer. Gene 659, 44–51. ( 10.1016/j.gene.2018.03.050) [DOI] [PubMed] [Google Scholar]
- 52.Slattery ML, Mullany LE, Sakoda L, Samowitz WS, Wolff RK, Stevens JR, Herrick JS. 2018. The NF-kappa B signalling pathway in colorectal cancer: associations between dysregulated gene and miRNA expression. J. Cancer Res. Clin. 144, 269–283. ( 10.1007/s00432-017-2548-6) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Liu JX, Liu F, Li X, Song X, Zhou L, Jie JZ. 2017. Screening key genes and miRNAs in early-stage colon adenocarcinoma by RNA-sequencing. Tumor Biol. 39, 1010428317714899 ( 10.1177/1010428317714899) [DOI] [PubMed] [Google Scholar]
- 54.Rammer M, et al. 2017. MicroRNAs and their role for T stage determination and lymph node metastasis in early colon carcinoma. Clin. Exp. Metastasis 34, 431–440. ( 10.1007/s10585-017-9863-9) [DOI] [PubMed] [Google Scholar]
- 55.Ozawa T, et al. 2018. A microRNA signature associated with metastasis of T1 colorectal cancers to lymph nodes. Gastroenterology 154, 844–848. ( 10.1053/j.gastro.2017.11.275) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Emami SS, Akbari A, Zare AA, Agah S, Masoodi M, Talebi A, Minaeian S, Fattahi A, Moghadamnia F. In press. MicroRNA expression levels and histopathological features of colorectal cancer. J. Gastrointest. Cancer. ( 10.1007/s12029-018-0055-x) [DOI] [PubMed] [Google Scholar]
- 57.Lundberg IV, Wikberg ML, Ljuslinder I, Li X, Myte R, Zingmark C, Lofgren-Burstrom A, Edin S, Palmqvist R. 2018. MicroRNA expression in KRAS- and BRAF-mutated colorectal cancers. Anticancer Res. 38, 677–683. ( 10.21873/anticanres.12272) [DOI] [PubMed] [Google Scholar]
- 58.Gomes SE, Pereira DM, Roma-Rodrigues C, Fernandes AR, Borralho PM, Rodrigues CMP. 2018. Convergence of miR-143 overexpression, oxidative stress and cell death in HCT116 human colon cancer cells. PLoS One 13, e0191607 ( 10.1371/journal.pone.0191607) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kim C, Hong Y, Lee H, Kang H, Lee EK. 2018. MicroRNA-195 desensitizes HCT116 human colon cancer cells to 5-fluorouracil. Cancer Lett. 412, 264–271. ( 10.1016/j.canlet.2017.10.022) [DOI] [PubMed] [Google Scholar]
- 60.Kong DF, Zhang DY, Chu XQ, Wang J. 2018. Schizandrin A enhances chemosensitivity of colon carcinoma cells to 5-fluorouracil through up-regulation of miR-195. Biomed. Pharmacother. 99, 176–183. ( 10.1016/j.biopha.2018.01.035) [DOI] [PubMed] [Google Scholar]
- 61.Wu N, Fesler A, Liu H, Ju J. 2018. Development of novel miR-129 mimics with enhanced efficacy to eliminate chemoresistant colon cancer stem cells. Oncotarget 9, 8887–8897. ( 10.18632/oncotarget.22322) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Marquez J, et al. 2018. Targeting liver sinusoidal endothelial cells with miR-20a-loaded nanoparticles reduces murine colon cancer metastasis to the liver. Int. J. Cancer. 143, 709–719. ( 10.1002/ijc.31343) [DOI] [PubMed] [Google Scholar]
- 63.Staedel C, Tran TPA, Giraud J, Darfeuille F, Di Giorgio A, Tourasse NJ, Salin F, Uriac P, Duca M. 2018. Modulation of oncogenic miRNA biogenesis using functionalized polyamines. Sci. Rep. 8, 1667 ( 10.1038/s41598-018-20053-5) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Zhang JX, et al. 2013. Prognostic and predictive value of a microRNA signature in stage II colon cancer: a microRNA expression analysis. Lancet Oncol. 14, 1295–1306. ( 10.1016/S1470-2045(13)70491-1) [DOI] [PubMed] [Google Scholar]
- 65.Huang SP, et al. 2014. Genetic variants in microRNAs and microRNA target sites predict biochemical recurrence after radical prostatectomy in localized prostate cancer. Int. J. Cancer 135, 2661–2667. ( 10.1002/ijc.28904) [DOI] [PubMed] [Google Scholar]
- 66.Grassi A, Perilli L, Albertoni L, Tessarollo S, Mescoli C, Urso EDL, Fassan M, Rugge M, Zanovello P. 2018. A coordinate deregulation of microRNAs expressed in mucosa adjacent to tumor predicts relapse after resection in localized colon cancer. Mol. Cancer 17, 17 ( 10.1186/s12943-018-0770-8) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Sazanov AA, Kiselyova EV, Zakharenko AA, Romanov MN, Zaraysky MI. 2017. Plasma and saliva miR-21 expression in colorectal cancer patients. J. Appl. Genet. 58, 231–237. ( 10.1007/s13353-016-0379-9) [DOI] [PubMed] [Google Scholar]
- 68.Toiyama Y, Takahashi M, Hur K, Nagasaka T, Tanaka K, Inoue Y, Kusunoki M, Boland CR, Goel A. 2013. Serum miR-21 as a diagnostic and prognostic biomarker in colorectal cancer. J. Natl Cancer Inst. 105, 849–859. ( 10.1093/jnci/djt101) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kijima T, et al. 2017. MicroRNA-6826 and-6875 in plasma are valuable non-invasive biomarkers that predict the efficacy of vaccine treatment against metastatic colorectal cancer. Oncol. Rep. 37, 23–30. ( 10.3892/or.2016.5267) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Maierthaler M, Benner A, Hoffmeister M, Surowy H, Jansen L, Knebel P, Chang-Claude J, Brenner H, Burwinkel B. 2017. Plasma miR-122 and miR-200 family are prognostic markers in colorectal cancer. Int. J. Cancer. 140, 176–187. ( 10.1002/ijc.30433) [DOI] [PubMed] [Google Scholar]
- 71.He HW, Wang NN, Yi XM, Tang CP, Wang D. 2018. Low-level serum miR-24-2 is associated with the progression of colorectal cancer. Cancer Biomark. 21, 261–267. ( 10.3233/Cbm-170321) [DOI] [PubMed] [Google Scholar]
- 72.Ng L, et al. 2017. Identification of serum miR-139-3p as a non-invasive biomarker for colorectal cancer. Oncotarget 8, 27 393–27 400. ( 10.18632/oncotarget.16171) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Miyoshi J, Toden S, Yoshida K, Toiyama Y, Alberts SR, Kusunoki M, Sinicrope FA, Goel A. 2017. MiR-139-5p as a novel serum biomarker for recurrence and metastasis in colorectal cancer. Sci. Rep. 7, 43393 ( 10.1038/srep43393) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Wang QJ, Zhang HC, Shen XJ, Ju SQ. 2017. Serum microRNA-135a-5p as an auxiliary diagnostic biomarker for colorectal cancer. Ann. Clin. Biochem. 54, 76–85. ( 10.1177/0004563216638108) [DOI] [PubMed] [Google Scholar]
- 75.Hur K, Toiyama Y, Okugawa Y, Ide S, Imaoka H, Boland CR, Goel A. 2017. Circulating microRNA-203 predicts prognosis and metastasis in human colorectal cancer. Gut 66, 654–664. ( 10.1136/gutjnl-2014-308737) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Liu HN, Liu TT, Wu H, Chen YJ, Tseng YJ, Yao C, Weng SQ, Dong L, Shen XZ. 2018. Serum microRNA signatures and metabolomics have high diagnostic value in colorectal cancer using two novel methods. Cancer Sci. 109, 1185–1194. ( 10.1111/cas.13514) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Fang ZX, et al. 2015. Plasma levels of microRNA-24, microRNA-320a, and microRNA-423-5p are potential biomarkers for colorectal carcinoma. J. Exp. Clin. Cancer Res. 34, 86 ( 10.1186/s13046-015-0198-6) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Tsukamoto M, Iinuma H, Yagi T, Matsuda K, Hashiguchi Y. 2017. Circulating exosomal microRNA-21 as a biomarker in each tumor stage of colorectal cancer. Oncology 92, 360–370. ( 10.1159/000463387) [DOI] [PubMed] [Google Scholar]
- 79.Yan SS, et al. 2018. Exosomal miR-6803-5p as potential diagnostic and prognostic marker in colorectal cancer. J. Cell. Biochem. 119, 4113–4119. ( 10.1002/jcb.26609) [DOI] [PubMed] [Google Scholar]
- 80.Fu FF, Jiang WQ, Zhou LF, Chen Z. 2018. Circulating exosomal miR-17-5p and miR-92a-3p predict pathologic stage and grade of colorectal cancer. Transl. Oncol. 11, 221–232. ( 10.1016/j.tranon.2017.12.012) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ghanbari R, Mosakhani N, Asadi J, Nouraee N, Mowla SJ, Poustchi H, Malekzadeh R, Knuutila S. 2015. Decreased expression of fecal miR-4478 and miR-1295b-3p in early-stage colorectal cancer. Cancer Biomark. 15, 189–195. ( 10.3233/Cbm-140453) [DOI] [PubMed] [Google Scholar]
- 82.Koberle V, et al. 2013. Differential stability of cell-free circulating microRNAs: implications for their utilization as biomarkers. PLoS One 8, e75184 ( 10.1371/journal.pone.0075184) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hall JS, Taylor J, Valentine HR, Irlam JJ, Eustace A, Hoskin PJ, Miller CJ, West CM. 2012. Enhanced stability of microRNA expression facilitates classification of FFPE tumour samples exhibiting near total mRNA degradation. Br. J. Cancer 107, 684–694. ( 10.1038/bjc.2012.294) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Gai Y, Wang HJ, Ma YY, Hu AX, Ma YL, Hu J, Yu YH. 2017. Serum miR-129 functions as a biomarker for colorectal cancer by targeting estrogen receptor (ER) beta. Pharmazie 72, 107–112. ( 10.1691/ph.2017.6718) [DOI] [PubMed] [Google Scholar]
- 85.Wang YN, Chen ZH, Chen WC. 2017. Novel circulating microRNAs expression profile in colon cancer: a pilot study. Eur. J. Med. Res. 22, 51 ( 10.1186/s40001-017-0294-5) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Link A, Balaguer F, Shen Y, Nagasaka T, Lozano JJ, Boland CR, Goel A. 2010. Fecal microRNAs as novel biomarkers for colon cancer screening. Cancer Epidem. Biomark. 19, 1766–1774. ( 10.1158/1055-9965.Epi-10-0027) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Zhu YX, Xu AD, Li JM, Fu JH, Wang GH, Yang YL, Cui L, Sun JW. 2016. Fecal miR-29a and miR-224 as the noninvasive biomarkers for colorectal cancer. Cancer Biomark. 16, 259–264. ( 10.3233/Cbm-150563) [DOI] [PubMed] [Google Scholar]
- 88.Chang PY, Chen CC, Chang YS, Tsai WS, You JF, Lin GP, Chen TW, Chen JS, Chan EC. 2016. MicroRNA-223 and microRNA-92a in stool and plasma samples act as complementary biomarkers to increase colorectal cancer detection. Oncotarget 7, 10 663–10 675. ( 10.18632/oncotarget.7119) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Koga Y, Yamazaki N, Yamamoto Y, Yamamoto S, Saito N, Kakugawa Y, Otake Y, Matsumoto M, Matsumura Y. 2013. Fecal miR-106a is a useful marker for colorectal cancer patients with false-negative results in immunochemical fecal occult blood test. Cancer Epidemiol. Biomarkers Prev. 22, 1844–1852. ( 10.1158/1055-9965.EPI-13-0512) [DOI] [PubMed] [Google Scholar]
- 90.Wu CW, et al. 2014. Identification of microRNA-135b in stool as a potential noninvasive biomarker for colorectal cancer and adenoma. Clin. Cancer Res. 20, 2994–3002. ( 10.1158/1078-0432.Ccr-13-1750) [DOI] [PubMed] [Google Scholar]
- 91.Bhome R, et al. 2017. Exosomal microRNAs derived from colorectal cancer-associated fibroblasts: role in driving cancer progression. Aging 9, 2666–2694. ( 10.18632/aging.101355) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Monzo M, Santasusagna S, Moreno I, Martinez F, Hernandez R, Munoz C, Castellano JJ, Moreno J, Navarro A. 2017. Exosomal microRNAs isolated from plasma of mesenteric veins linked to liver metastases in resected patients with colon cancer. Oncotarget 8, 30 859–30 869. ( 10.18632/oncotarget.16103) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Hosseini M, Khatamianfar S, Hassanian SM, Nedaeinia R, Shafiee M, Maftouh M, Ghayour-Mobarhan M, ShahidSales S, Avan A. 2017. Exosome-encapsulated microRNAs as potential circulating biomarkers in colon cancer. Curr. Pharm. Design 23, 1705–1709. ( 10.2174/1381612822666161201144634) [DOI] [PubMed] [Google Scholar]
- 94.Clancy C, et al. 2016. Screening of exosomal microRNAs from colorectal cancer cells. Cancer Biomark. 17, 427–435. ( 10.3233/Cbm-160659) [DOI] [PubMed] [Google Scholar]
- 95.Uratani R, et al. 2016. Diagnostic potential of cell-free and exosomal microRNAs in the identification of patients with high-risk colorectal adenomas. PLoS ONE 11, e0160722 ( 10.1371/journal.pone.0160722) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Tovar-Camargo OA, Toden S, Goel A. 2016. Exosomal microRNA biomarkers: emerging frontiers in colorectal and other human cancers. Expert Rev. Mol. Diagn. 16, 553–567. ( 10.1586/14737159.2016.1156535) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Roberts BS, et al. 2018. Discovery and validation of circulating biomarkers of colorectal adenoma by high-depth small RNA sequencing. Clin. Cancer Res. 24, 2092–2099. ( 10.1158/1078-0432.Ccr-17-1960) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Melo SA, et al. 2014. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell. 26, 707–721. ( 10.1016/j.ccell.2014.09.005) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Okoye IS, Coomes SM, Pelly VS, Czieso S, Papayannopoulos V, Tolmachova T, Seabra MC, Wilson MS. 2014. MicroRNA-containing T-regulatory-cell-derived exosomes suppress pathogenic T helper 1 cells. Immunity 41, 89–103. ( 10.1016/j.immuni.2014.05.019) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.O'Driscoll L. 2015. Expanding on exosomes and ectosomes in cancer. N. Engl. J. Med. 372, 2359–2362. ( 10.1056/NEJMcibr1503100) [DOI] [PubMed] [Google Scholar]
- 101.Matsumura T, et al. 2015. Exosomal microRNA in serum is a novel biomarker of recurrence in human colorectal cancer. Br. J. Cancer 113, 275–281. ( 10.1038/bjc.2015.201) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Ogata-Kawata H, et al. 2014. Circulating exosomal microRNAs as biomarkers of colon cancer. PLoS ONE 9, e92921 ( 10.1371/journal.pone.0092921) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Liu C, et al. 2016. Serum exosomal miR-4772-3p is a predictor of tumor recurrence in stage II and III colon cancer. Oncotarget 7, 76 250–76 260. ( 10.18632/oncotarget.12841) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Baxter NN, Warren JL, Barrett MJ, Stukel TA, Doria-Rose VP. 2012. Association between colonoscopy and colorectal cancer mortality in a US cohort according to site of cancer and colonoscopist specialty. J. Clin. Oncol. 30, 2664–2669. ( 10.1200/Jco.2011.40.4772) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Baxter NN, Goldwasser MA, Paszat LF, Saskin R, Urbach DR, Rabeneck L. 2009. Association of colonoscopy and death from colorectal cancer. Ann. Intern. Med. 150, 1–8. ( 10.7326/0003-4819-150-1-200901060-00306) [DOI] [PubMed] [Google Scholar]
- 106.Nicholson FB, Korman MG. 2005. Acceptance of flexible sigmoidoscopy and colonoscopy for screening and surveillance in colorectal cancer prevention. J. Med. Screen. 12, 89–95. ( 10.1258/0969141053908294) [DOI] [PubMed] [Google Scholar]
- 107.Gili M, Roca M, Ferrer V, Obrador A, Cabeza E. 2006. Psychosocial factors associated with the adherence to a colorectal cancer screening program. Cancer Detect. Prev. 30, 354–360. ( 10.1016/j.cdp.2006.06.005) [DOI] [PubMed] [Google Scholar]
- 108.Duffy MJ, van Dalen A, Haglund C, Hansson L, Klapdor R, Lamerz R, Nilsson O, Sturgeon C, Topolcan O. 2003. Clinical utility of biochemical markers in colorectal cancer: European Group on Tumour Markers (EGTM) guidelines. Eur. J. Cancer 39, 718–727. ( 10.1016/S0959-8049(02)00811-0) [DOI] [PubMed] [Google Scholar]
- 109.Levin B, et al. 2008. Screening and surveillance for the early detection of colorectal cancer and adenomatous polyps, 2008: a joint guideline from the American Cancer Society, the US Multi-Society Task Force on Colorectal Cancer, and the American College of Radiology. CA Cancer J. Clin. 58, 130–160. ( 10.3322/Ca.2007.0018) [DOI] [PubMed] [Google Scholar]
- 110.Guo SH, Zhang JJ, Wang BS, Zhang BD, Wang XN, Huang L, Liu HY, Jia BQ. 2018. A 5-serum miRNA panel for the early detection of colorectal cancer. Oncotargets Ther. 11, 2603–2614. ( 10.2147/Ott.S153535) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Wang SY, et al. 2015. A plasma microRNA panel for early detection of colorectal cancer. Int. J. Cancer 136, 152–161. ( 10.1002/ijc.28136) [DOI] [PubMed] [Google Scholar]
- 112.Shidfar A, et al. 2017. Expression of miR-18a and miR-210 in normal breast tissue as candidate biomarkers of breast cancer risk. Cancer Prev. Res. 10, 89–97. ( 10.1158/1940-6207.CAPR-16-0177) [DOI] [PubMed] [Google Scholar]
- 113.Xu X, Zhu S, Tao Z, Ye S. 2018. High circulating miR-18a, miR-20a, and miR-92a expression correlates with poor prognosis in patients with non-small cell lung cancer. Cancer Med. 7, 21–31. ( 10.1002/cam4.1238) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Zuo J, Yu Y, Zhu M, Jing W, Yu M, Chai H, Liang C, Tu J. 2018. Inhibition of miR-155, a therapeutic target for breast cancer, prevented in cancer stem cell formation. Cancer Biomark. 21, 383–392. ( 10.3233/CBM-170642) [DOI] [PubMed] [Google Scholar]
- 115.Park S, et al. 2017. MiR-9, miR-21, and miR-155 as potential biomarkers for HPV positive and negative cervical cancer. BMC Cancer 17, 658 ( 10.1186/s12885-017-3642-5) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Carter JV, Galbraith NJ, Yang D, Burton JF, Walker SP, Galandiuk S. 2017. Blood-based microRNAs as biomarkers for the diagnosis of colorectal cancer: a systematic review and meta-analysis. Br. J. Cancer 116, 762–774. ( 10.1038/bjc.2017.12) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Carthew RW, Sontheimer EJ. 2009. Origins and mechanisms of miRNAs and siRNAs. Cell 136, 642–655. ( 10.1016/j.cell.2009.01.035) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Slezak-Prochazka I, Durmus S, Kroesen BJ, van den Berg A. 2010. MicroRNAs, macrocontrol: regulation of miRNA processing. RNA 16, 1087–1095. ( 10.1261/rna.1804410) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Kim B, Jeong K, Kim VN. 2017. Genome-wide mapping of DROSHA cleavage sites on primary microRNAs and noncanonical substrates. Mol. Cell 66, 258–269. ( 10.1016/j.molcel.2017.03.013) [DOI] [PubMed] [Google Scholar]
- 120.Varol D, et al. 2017. Dicer deficiency differentially impacts microglia of the developing and adult brain. Immunity 46, 1030–1044. ( 10.1016/j.immuni.2017.05.003) [DOI] [PubMed] [Google Scholar]
- 121.Sun HL, et al. 2016. ERK activation globally downregulates miRNAs through phosphorylating exportin-5. Cancer Cell 30, 723–736. ( 10.1016/j.ccell.2016.10.001) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Reddy KB. 2015. MicroRNA (miRNA) in cancer. Cancer Cell Int. 15, 38 ( 10.1186/s12935-015-0185-1) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Lujambio A, et al. 2008. A microRNA DNA methylation signature for human cancer metastasis. Proc. Natl Acad. Sci. USA 105, 13 556–13 561. ( 10.1073/pnas.0803055105) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Bandres E, Agirre X, Bitarte N, Ramirez N, Zarate R, Roman-Gomez J, Prosper F, Garcia-Foncillas J. 2009. Epigenetic regulation of microRNA expression in colorectal cancer. Int. J. Cancer 125, 2737–2743. ( 10.1002/ijc.24638) [DOI] [PubMed] [Google Scholar]
- 125.Dai DJ, Wang HY, Zhu LY, Jin HC, Wang X. 2018. N6-methyladenosine links RNA metabolism to cancer progression. Cell Death Dis. 9, 124 ( 10.1038/s41419-017-0129-x) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Otsuka K, Yamamoto Y, Matsuoka R, Ochiya T. 2018. Maintaining good miRNAs in the body keeps the doctor away?: perspectives on the relationship between food-derived natural products and microRNAs in relation to exosomes/extracellular vesicles. Mol. Nutr. Food Res. 62, 1700080 ( 10.1002/mnfr.201700080) [DOI] [PubMed] [Google Scholar]
- 127.Nishida-Aoki N, Ochiya T. 2015. Interactions between cancer cells and normal cells via miRNAs in extracellular vesicles. Cell. Mol. Life Sci. 72, 1849–1861. ( 10.1007/s00018-014-1811-0) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Kosaka N, Ochiya T. 2011. Unraveling the mystery of cancer by secretory microRNA: horizontal microRNA transfer between living cells. Front. Genet. 2, 97 ( 10.3389/fgene.2011.00097) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, Matsuki Y, Ochiya T. 2010. Secretory mechanisms and intercellular transfer of microRNAs in living cells. J. Biol. Chem. 285, 17 442–17 452. ( 10.1074/jbc.M110.107821) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Wei JX, et al. 2015. Vps4A functions as a tumor suppressor by regulating the secretion and uptake of exosomal microRNAs in human hepatoma cells. Hepatology 61, 1284–1294. ( 10.1002/hep.27660) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Jae N, McEwan DG, Manavski Y, Boon RA, Dimmeler S. 2015. Rab7a and Rab27b control secretion of endothelial microRNA through extracellular vesicles. FEBS Lett. 589, 3182–3188. ( 10.1016/j.febslet.2015.08.040) [DOI] [PubMed] [Google Scholar]
- 132.Moody L, He HS, Pan YX, Chen H. 2017. Methods and novel technology for microRNA quantification in colorectal cancer screening. Clin. Epigenet. 9, 119 ( 10.1186/s13148-017-0420-9) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Kesici E, Eksin E, Erdem A. 2018. An impedimetric biosensor based on ionic liquid-modified graphite electrodes developed for microRNA-34a detection. Sensors 18, 2868 ( 10.3390/s18092868) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Zhang N, Shi XM, Guo HQ, Zhao XZ, Zhao WW, Xu JJ, Chen HY. 2018. Gold nanoparticle couples with entropy-driven toehold-mediated DNA strand displacement reaction on magnetic beads: toward ultrasensitive energy-transfer-based photoelectrochemical detection of miRNA-141 in real blood sample. Anal. Chem. 90, 11 892–11 898. ( 10.1021/acs.analchem.8b01966) [DOI] [PubMed] [Google Scholar]
- 135.Tian L, Qi J, Ma X, Wang X, Yao C, Song W, Wang Y. 2018. A facile DNA strand displacement reaction sensing strategy of electrochemical biosensor based on N-carboxymethyl chitosan/molybdenum carbide nanocomposite for microRNA-21 detection. Biosens. Bioelectron. 122, 43–50. ( 10.1016/j.bios.2018.09.037) [DOI] [PubMed] [Google Scholar]
- 136.Liu X, Zhang SQ, Cheng ZH, Wei X, Yang T, Yu YL, Chen ML, Wang JH. 2018. Highly sensitive detection of microRNA-21 with ICPMS via hybridization accumulation of upconversion nanoparticles. Anal. Chem. 90, 12 116–12 122. ( 10.1021/acs.analchem.8b03038) [DOI] [PubMed] [Google Scholar]
- 137.Liu Q, Fan J, Zhou C, Wang L, Zhao B, Zhang H, Liu B, Tong C. 2018. Quantitative detection of miRNA-21 expression in tumor cells and tissues based on molecular beacon. Int. J. Anal. Chem. 2018, 3625823 ( 10.1155/2018/3625823) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Boriachek K, Umer M, Islam MN, Gopalan V, Lam AK, Nguyen NT, Shiddiky MJA. 2018. An amplification-free electrochemical detection of exosomal miRNA-21 in serum samples. Analyst 143, 1662–1669. ( 10.1039/c7an01843f) [DOI] [PubMed] [Google Scholar]
- 139.Bell E, Watson HL, Bailey S, Murray MJ, Coleman N. 2017. A robust protocol to quantify circulating cancer biomarker microRNAs. Methods Mol. Biol. 1580, 265–279. ( 10.1007/978-1-4939-6866-4_18) [DOI] [PubMed] [Google Scholar]
- 140.Kai K, Dittmar RL, Sen S. 2018. Secretory microRNAs as biomarkers of cancer. Semin. Cell Dev. Biol. 78, 22–36. ( 10.1016/j.semcdb.2017.12.011) [DOI] [PubMed] [Google Scholar]
- 141.Fujii S, Kamiya K, Osaki T, Misawa N, Hayakawa M, Takeuchi S. 2018. Purification-free microRNA detection by using magnetically immobilized nanopores on liposome membrane. Anal. Chem. 90, 10 217–10 222. ( 10.1021/acs.analchem.8b01443) [DOI] [PubMed] [Google Scholar]
- 142.Lu W, Wang Y, Song S, Chen C, Yao B, Wang M. 2018. A fishhook probe-based rolling circle amplification (FP-RCA) assay for efficient isolation and detection of microRNA without total RNA extraction. Analyst 143, 5046–5053. ( 10.1039/c8an01544a) [DOI] [PubMed] [Google Scholar]
- 143.Stanitz E, Juhasz K, Gombos K, Gocze K, Toth C, Kiss I. 2015. Alteration of miRNA expression correlates with lifestyle, social and environmental determinants in esophageal carcinoma. Anticancer Res. 35, 1091–1097. [PubMed] [Google Scholar]
- 144.Stanitz E, Juhasz K, Toth C, Gombos K, Natali PG, Ember I. 2013. Evaluation of microRNA expression pattern of gastric adenocarcinoma associated with socioeconomic, environmental and lifestyle factors in northwestern Hungary. Anticancer Res. 33, 3195–3200. [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
This article has no additional data.