Open access peer-reviewed chapter

MicroRNA in Inflammatory Bowel Disease

Written By

Kurt Fisher and Jingmei Lin

Submitted: 20 October 2015 Reviewed: 07 July 2016 Published: 26 October 2016

DOI: 10.5772/64836

From the Edited Volume

New Insights into Inflammatory Bowel Disease

Edited by Samuel Huber

Chapter metrics overview

1,853 Chapter Downloads

View Full Metrics

Abstract

Idiopathic inflammatory bowel disease (IBD) is a complex set of disorders that predominantly includes ulcerative colitis and Crohn’s disease. The pathogenesis of IBD is multifactorial including genetic, infectious, and immunologic factors. MicroRNAs belong to a class of noncoding small RNAs that posttranscriptionally regulate gene expression, and they are an emerging class of genetic modifiers of IBD. Here, we focus on the use of unique microRNA expression patterns as biomarkers to classify and prognosticate disease severity in both mucosal tissue and serum from patients with either ulcerative colitis or Crohn’s disease. Furthermore, we discuss specific microRNAs with respect to their roles in IBD pathogenesis and fibrosis. We also discuss the role of microRNAs in IBD-associated carcinogenesis, including their role as biomarkers, tumor suppressors, and oncogenes. Finally, we discuss the emerging therapeutic applications of microRNA manipulation to lessen the effect of IBD and its sequelae. Recent discoveries of the diverse roles of microRNAs in IBD pathogenesis have the potential to provide new targeted therapeutics for personalized medicine.

Keywords

  • inflammatory bowel disease
  • ulcerative colitis
  • Crohn’s disease
  • microRNA
  • pathogenesis

1. Introduction

Idiopathic inflammatory bowel disease (IBD) is a group of chronic and recurrent inflammatory disorders that primarily involve the gastrointestinal tract. It predominantly includes ulcerative colitis (UC) and Crohn’s disease (CD). The pathogenesis of IBD is multifactorial and not completely understood, but genetic, epigenetic, infectious, physiological, and immunological factors may all play important roles in the genesis and progression of the diseases [13]. A large number of genes have been linked to IBD susceptibility, pathogenesis, and carcinogenesis, and recent work has suggested that microRNAs can undertake the same roles.

MicroRNAs are encoded within the genomes of a wide variety of eukaryotes, and more than 2500 human mature microRNAs have been curated in miRBase database since their discovery in 1993 [4, 5]. MicroRNAs are evolutionarily conserved, single-stranded noncoding RNA molecules of 19–24 nucleotides, which represent a class of regulatory RNAs suppressing gene expression at a posttranscriptional level. MicroRNAs concurrently modulate the expression levels of dozens or more messenger RNA (mRNA) targets and any given mRNA sequence may be targeted by several different microRNAs creating intricate regulatory networks to fine-tune a cell’s function [68]. At the time of publication, microRNAs are predicted to directly regulate the expression of at least 30% of the entire mammalian genome [9]. MicroRNAs have been found to be involved in the normal functioning of multiple pathophysiological networks and in the pathogenesis of a broad spectrum of human diseases, ranging from neoplastic to inflammatory conditions [1015].

In this chapter, we focus on the role of microRNAs in IBD as recent publications have indicated that microRNAs play critical roles in the pathogenesis of IBD and IBD-associated carcinogenesis and may serve as critical future targets for personalized medicine.

Advertisement

2. MicroRNAs show a diverse array of aberrant expression in IBD

Extensive literature has shown that microRNAs undergo dysregulation in both tissue and peripheral blood of patients with IBD with a goal of discovering biomarkers and crucial initiators of pathogenesis.

2.1. MicroRNAs are differentially expressed in the mucosal tissue in UC

A search for biomarkers in UC in both non-affected and actively inflamed mucosa has identified a large number of microRNAs that show aberrant expression (Table 1) [1628]. When compared to controls, a large number of microRNAs have been found to be upregulated including, but not limited to, miR-let-7e*, miR-let-7f, miR-7, miR-7i, miR-16, miR-20b, miR-21, miR-23a, miR-24, miR-29a, miR-29b, miR-31, miR-98, miR-125b-1*, miR-126, miR-126*, miR-127-3p, miR-135b, miR-142-3p, miR-146a, miR-150, miR-155, miR-195, miR-196a, miR-206, miR-223, miR-324-3p, miR-375, miR-422b, miR-548a-3p, miR-650, miR-663, and miR-4284. The decreased microRNA profiles include miR-143, miR-145, miR-188-5p, miR-192, miR-194, miR-194b, miR-196b, miR-200b, miR-215, miR-216b, miR-320a, miR-346, miR-375, miR-489, miR-548e, miR-559, and miR-630. Seven microRNA candidates have been identified by at least two independent groups, miR-21 [1618, 26, 27], miR-29a [16, 19], miR-31 [19, 24], miR-146a [23, 24, 27], miR-155 [17, 23], miR-192 [16, 27], and miR-375 [16, 27], making them the most promising candidates for biomarkers of UC.

However, there are many variables that differ between studies, including anatomical tissue location, degree of inflammation, prior medication, and platforms used to measure microRNAs that all may contribute to the high discordance between studies.

Status Tissue type Control Aberrant microRNA expression Reference
Active UC Sigmoid, n = 15 Healthy Decreased: miR-192 and 375
Increased: miR-let-7f, 16, 21, 23a, 24, 29a, 126, 195, and 422b
Wu [16]
Sigmoid, n = 12 Healthy Increased: miR-21 and 155 Takagi [17]
Sigmoid, n = 12 Healthy Increased: miR-21 and 126 Feng [18]
Colon, nonspecific, n = 10 Healthy Decreased: miR-188-5p, 215, 320a, and 346
Increased: miR-7, 31, 135b, and 223
Fasseu [19]
Colon, nonspecific, n = 5 Healthy Increased: miR-150 Bian [20]
Colon, nonspecific, n = 8 Healthy Decreased: miR-143 and 145 Pekow [21]
Colon, nonspecific, n = 20 Healthy Increased: miR-let-7e, 20b, and 98* Coskun [22]
Colon, n = 10 Healthy Increased: miR-146a and 155 Beres [23]
Active or inactive UC Colon, distal-most, n = 10 Healthy Decreased: miR-194b, 216b, 548e, and 559
Increased: miR-31, 146a, 206, and 663
Lin [24]
Sigmoid, n = 26 Healthy Increased: miR-4284 Koukos [25]
Inactive UC Sigmoid, n = 15 Healthy Increased: miR-16, 23a, 24, 29a, 375, and 422b  Wu [16]
Colon, nonspecific, n = 8 Healthy Decreased: miR-188-5p, 215, 320a, and 346
Increased: miR-29a, 29b, 126*, 127-3p, 196a, and 324-3p
Fasseu [19]
Colon, nonspecific, n = 19 Healthy Increased: miR-20b and 125b-1* Coskun [22]
Unknown Colon, nonspecific, n = 15 Healthy Increased: miR-21 Yang [26]
Colon, nonspecific, n = 12 Healthy Increased: miR-7i, 21, 142-3p, and 146a
Decreased: miR-192, 194, 200b, and 375
Zahm [27]
Active UC Colon, nonspecific, n = 20 Inactive UC  Increased: miR-98 Coskun [22]
Colon, left or sigmoid, n = 9  Inactive UC Decreased: miR-196b, 489, and 630
Increased: miR-548a-3p and 650
Iborra [28]

Table 1.

Aberrant microRNA expression in human colonic tissue in ulcerative colitis (UC).

2.2. MicroRNAs are differentially expressed in the mucosal tissue in CD

A search for biomarkers in CD has identified a large number of microRNAs that show aberrant expression (Table 2) [19, 23, 24, 2832]. When compared to controls, miR-9, miR-9*, miR-16, miR-21, miR-22, miR-23b, miR-26a, miR-29b, miR-29c, miR-30a, miR-30b, miR-30c, miR-31, miR-34c-5p, miR-106a, miR-122, miR-126, miR-126*, miR-127-3p, miR-130a, miR-133b, miR-141, miR-146a, miR-146b-5p, miR-150, miR-155, miR-181c, miR-191, miR-196, miR-196a, miR-206, miR-223, miR-324-3p, miR-328, miR-375, miR-422a, miR-594, miR-663, and miR-885-5p have been found significantly upregulated [19, 24, 29, 31, 32]. The downregulated microRNA profiles include miR-let-7b, miR-7, miR-18a*, miR-19b, miR140-3p, miR-194b, miR-216b, miR-548e, miR-559, miR-629, and miR-629* [24, 30, 33].

Status Tissue type Control  Aberrant microRNA expression Reference
Active CD Sigmoid, n = 5 Healthy Decreased: miR-19b and 629
Increased: miR-23b, 106a, and 191
Wu [29]
Terminal ileum, n = 6 Healthy Increased: miR-16, 21, 223, and 594 Wu [29]
Colon, nonspecific, n = 16 Healthy Increased: miR-9, 21, 22, 26a, 29b, 29c, 30b, 31,
34c-5p, 106a, 126, 126*, 127-3p, 130a, 133b,
146a, 146b-5p, 150, 155, 181c, 196a, 324-3p,
and 375
Fasseu [19]
Colon, nonspecific, n = 8 Healthy Decreased: miR-7 Nguyen [30]
Colon, nonspecific, n = 120  Healthy Increased: miR-196 Brest [31]
Colon, nonspecific, n = 15 Healthy Increased: miR-31 and 141 Huang [32]
Colon, nonspecific, n = 10 Healthy Increased: miR-146a and 155 Beres [23]
Active and inactive CD Colon, distal-most, n = 9 Healthy Decreased: miR-194b, 216b, 548e, and 559
Increased: miR-31, 146a, 206, and 663
Lin [24]
Inactive CD Colon, nonspecific, n = 8 Healthy Increased: miR-9*, 21, 22, 26a, 29b, 29c, 30a*, 30b, 30c, 31, 34c-5p, 106a, 126*, 127-3p, 133b, 146a, 146b-5p, 150, 155, 196a, 223, and 324-3p Fasseu [19]
Unknown Colon, nonspecific, n = 10 Healthy Increased: miR-122 Beres [23]
Colon, nonspecific, n = 7 Healthy Increased: miR-21 and 375 Zahm [27]
Active CD Colon, left or sigmoid, n = 9  Inactive
CD
Decreased: miR-let-7b,18a*, 140-3p, and 629*
Increased: miR-328, 422a, and 885-5p
Iborra [28]

Table 2.

Aberrant microRNA expression in human colonic tissue in Crohn’s disease (CD).

At least two independent groups have found similar dysregulation of miR-21 [19, 27, 29], miR-31 [19, 24, 32], miR-106a [19, 29], miR-146a [19, 23, 24], miR-155 [19, 23], miR-223 [19, 29], and miR-375 [19, 27].

2.3. MicroRNAs are differentially expressed in blood samples in UC

Similar to the findings in tissue, microRNAs are also dysregulated in the peripheral blood of patients with UC (Table 3) [26, 28, 3439]. When compared to controls, miR-let-7d, miR-let-7e, miR-let-7g, miR-let-7i*, miR-plus-E1271, miR-15b, miR-16, miR-17-5p, miR-19a, miR-20b*, miR-21, miR-22, miR-22-3p, miR-23a-3p, miR-24, miR-27a*, miR-28-3p, miR-28-5p, miR-29a, miR-30e, miR-30e-5p, miR-31, miR-92a-1*, miR-93, miR-103, miR-103-2, miR-103-2*, miR-128, miR-138, miR-140-3p, miR-142-5p, miR-143*, miR-146a-3p, miR-148b-3p, miR-150*, miR-151-5p, miR-155, miR-181b, miR-188-5p, miR-191-5p, miR-196b, miR-199a-3p, miR-199a-5p, miR-221, miR-223, miR-223a-3p, miR-302-3p, miR-320e, miR-330-3p, miR-340*, miR-345, miR-362-3p, miR-362-5p, miR-374b, miR-378, miR-378*, miR-422a, miR-423-5p, miR-500, miR-501-5p, miR-532-3p, miR-532-5p, miR-550*, miR-595, miR-598, miR-720, miR-760, miR-769-3p, miR-769-5p, miR-874, miR-941, miR-1246, miR-1271, miR-1274b, miR-1296, and miR-4516 are upregulated in peripheral blood samples of patients with UC [26, 28, 34, 35, 38, 39]. When compared to controls, miR-150, miR-188-5p, miR-505*, miR-612, and miR-1827 have been shown to be downregulated [28, 34, 38].

Status Tissue type Control Aberrant microRNA expression Reference
Active UC Peripheral blood, n = 13  Healthy Decreased: miR-505*
Increased: miR-plus-E1271, 28-5p, 103-2*,
151-5p, 199a-5p, 340*, 362-3p, and 532-3p
Wu [34]
Peripheral blood, n = 88 Healthy Increased: miR-16, 21, 28-5p, 151-5p, 155, and 199a-5p Paraskevi [35]
Peripheral blood, n = 62 Healthy Increased: miR-595 and 1246 Krissansen [36]
Active and inactive UC Peripheral blood, n = 18 Healthy Decreased: miR-150
Increased: miR-let-7d, let-7e, let-7g, 15b, 19a,
24, 27a, 28-3p, 29a, 30e, 93, 103, 128, 142-5p,
196b, 199a-3p, 221, 223, 345, 374b, 423-5P,
532-5p, 598, and 760
Iborra [28]
Peripheral blood, n = 46 Healthy Decreased: miR-188-5p, 612, and 1827
Increased: miR-17-5p, 22-3p, 23a-3p, 30e-5p, 148b-3p, 191-5p, 223a-3p, 302-3p, and 320e
Polytarchou [37]
Active UC Peripheral blood, n = 24 Inactive UC Increased: miR-23a-3p, 148b-3p, 320e, and
4516
Polytarchou [37]
Inactive UC Peripheral blood, n = 13 Healthy Decreased: miR-505*
Increased: miR-103-2, 362-3p, and 532-3p
Zahm [38]
Peripheral blood, n = 10 Healthy Decreased: miR-505*
Increased: miR-103-2*, 362-3p, and 532-3p
Wu [34]
Unknown Peripheral blood, n = 20 Healthy Increased: miR-let-7i*, 20b*, 22, 27a*, 31, 92a-1*, 138, 140-3p, 143*, 146a-3p, 150*,181b, 188-5p, 330-3p, 362-5p, 345, 378, 378*, 422a, 500,
501-5p, 532-5p, 550*, 720, 769-3p, 769-5p, 874, 941,
1271, 1274b, and 1296
Duttagupta [39]
Peripheral blood, n = 15 Healthy Increased: miR-21 Yang [26]

Table 3.

Aberrant microRNA expression in human peripheral blood in ulcerative colitis (UC).

Nine microRNAs have been found by at least two independent groups, miR-21 [26, 35], miR-28-5p [34, 35], miR-151-5p [34, 35], miR-199a-5p [34, 35], miR-345 [28, 39], miR-362-3p [34, 38], miR-505* [34, 38], miR-532-3p [34, 38], and miR-532-5p [28, 39].

2.4. MicroRNAs are differentially expressed in blood samples in CD

Similar to the findings in tissue, microRNAs are also dysregulated in the peripheral blood of patients with CD (Table 4) [28, 3436, 38]. When compared to controls, a large number of microRNAs have been found to be upregulated in the peripheral blood including miR-plus-E1271, miR-let-7b, miR-16, miR-20a, miR-21, miR-23a, miR-27a*, miR-29a, miR-30e, miR-93, miR-106a, miR-107, miR-126, miR-140, miR-140-3p, miR-140-5p, miR-188-5p, miR-191, miR-192, miR-195, miR-199a-5p, miR-200c, miR-340*, miR-362-3p, miR-484, miR-532-3p, miR-595, miR-877, and miR-1246. The significantly decreased microRNAs consist of miR-plus-F1065, miR-18a, miR-128, miR-140-5p, miR-145, miR-149*, and miR-877.

Status Tissue type Control  Aberrant microRNA expression Reference
Active CD Peripheral blood, n = 14  Healthy Decreased: miR-plus-F1065 and 149*
Increased: miR-plus-E1271, 199a-5p, 340*,
362-3p, and 532-3p
Wu [34]
Peripheral blood, n = 46 Healthy  Increased: miR-let-7b,16, 20a, 21, 30e, 93, 106a, 140, 192, 195, and 484 Zahm [38]
Peripheral blood, n = 128 Healthy Increased: miR-16, 23a, 29a, 106a, 107, 126, 191, 199a-5p, 200c, 362-3p, and 532-3p Paraskevi [35]
Peripheral blood, n = 57 Healthy Increased: miR-595 and 1246 Krissansen [36]
Active and inactive CD Peripheral blood, n = 18 Healthy Decreased: miR-877
Increased: miR-16, 27a*, 140-3p, 140-5p,
and 195
Iborra [28]
Inactive CD Peripheral blood, n = 5 Healthy Decreased: miR-149*
Increased: miR-340*
Wu [34]
Active CD Peripheral blood, n = 9 Inactive
CD
Decreased: miR-18a, 128, 140-5p, and 145
Increased: miR-188-5p and 877
Iborra [28]

Table 4.

Aberrant microRNA expression in human peripheral blood in Crohn’s disease (CD).

Six microRNAs have been found by at least two independent groups including miR-16 [28, 35, 38], miR-106a [35, 38], miR-195 [28, 38], miR-199a-5p [34, 35], miR-362-3p [34, 35], and miR-532-3p [34, 35].

2.5. Differential expression of microRNA that distinguishes UC from CD in indeterminate IBD

Although UC and CD have many overlapping features and symptomatology, they have distinct clinical, radiographic, endoscopic, surgical, and histologic findings. While most patients can be definitively classified as either UC or CD, 5–10% of IBD patients have equivocal features and are best classified as indeterminate colitis [40, 41]. The ability to determine which patients with indeterminate colitis act like CD would allow for better clinical and surgical management because patients with CD are much more likely to fail a pouch procedure.

Several microRNAs have been found to be differentially expressed between UC and CD, although the results have been inconsistent between most studies (Table 5) [19, 27, 34, 42].

In one study, a panel of five microRNAs (miR-19b, miR-23b, miR-106a, miR-191, and miR-629) was evaluated in 16 patients with a clinical diagnosis of indeterminate colitis. They found that 15 of 16 patients demonstrated UC-like expression patterns and concluded that microRNA expression patterns in indeterminate colitis are far more similar to those of UC than CD [42]. These microRNA expression findings are similar to the data from long-term clinical follow-ups where most indeterminate colitis patients act much more like UC patients, rather than CD patients. The possibility to test microRNA profiles prior to surgery, with the hope of improving pouch outcome, is promising.

Status Tissue type Control Aberrant microRNA expression Reference
Inactive UC Colon, nonspecific,
n = 8
Inactive CD Decreased: miR-100a-3p, 100b-5p,
150, 196b, 223, and 320a
Fasseu [19]
Active and inactive
UC
Colon, distal-most,
n = 12
Active and inactive
CD
Increased: miR-19b, 23b, 106a, 191,
and 629
Lin [42]
Unknown UC Colon, nonspecific,
n = 12
Unknown CD Increased: miR-24 Zahm [27]
Active UC Peripheral blood,
n = 13
Active CD Increased: miR-plus-E1035,
plus-F1159, and 3180-3p,
Wu [34]

Table 5.

Differential microRNA expression between ulcerative colitis (UC) and Crohn’s disease (CD).

Advertisement

3. MicroRNA as a potential driver of pathogenesis and disease severity

Despite the vast numbers of microRNAs identified as deregulated in IBD, very few microRNAs are replicated in multiple studies. Here, we focus on the microRNAs with the best evidence as the driver of pathogenesis.

3.1. MiR-21 has an essential role in IBD development and disease severity

MiR-21 has been consistently identified as being upregulated in active UC and CD, suggesting a central role in the pathogenesis of IBD [1618, 26, 43]. One study showed that the deletion of critical DNA methyltransferases (DNMT1 and DNMT3b) caused the dysregulation of approximately 10% of all microRNAs, highlighting the epigenetic regulation of microRNAs by DNA methylation [44]. The genome-wide association studies (GWASs) using Illumina CpG methylation assays have shown that the miR-21 locus was hypomethylated, and subsequently overexpressed, in samples of peripheral blood in patients with active CD [43]. A miR-21 knockout mouse model has been developed to assess the importance of miR-21 in IBD pathogenesis. Similar to human IBD, mice treated with dextran sodium sulfate (DSS) developed a chronic colitis causing significant morbidity and mortality with elevated tumor necrosis factor alpha (TNF-α) levels and miR-21 [45]. However, mice with genetic deletion of miR-21 were resistant to DSS-induced colitis. These findings strongly support the role of miR-21 in IBD pathogenesis, but further work is needed to clarify the mechanism of action.

The pathogenic effects of miR-21 in IBD are thought to be mediated through at least three separate mechanisms. First, miR-21 is thought to cause increased intestinal permeability in response to epithelial damage, a factor thought to initiate IBD. Despite no difference at baseline, treatment with DSS caused increased intestinal permeability in wild-type mice compared to miR-21 knockout mice [45]. Second, miR-21 is proapoptotic as the DSS-treated miR-21 knockout mice had less intestinal epithelial cell apoptosis than controls [45]. Several studies have indicated a role for miR-21 in the protection of free radical-induced apoptosis that linked the pro-survival phenotype to the inhibition of phosphatase and tensin homology (PTEN) with subsequent elevation of PI3K-Akt-mTOR activity [4649]. Additionally, miR-21 has been shown to prevent renal tubular apoptosis by directly reducing levels of Rab11 protein [50]. The prevention of epithelial cell apoptosis may help maintain the epithelial cell barrier and limit intestinal permeability. Third, miR-21 has been associated with fibrosis in multiple disease models and has an emerging role in irreversible fibrosis of IBD. Multiple models of cellular injury have shown to be dependent on increased levels of TNF-α and subsequent induction of miR-21 [51, 52]. Increased serum levels of miR-21 were seen in human diseases with significant fibrosis, suggesting a role for miR-21 as a biomarker for disease activity [52, 53]. Elevated miR-21 expression is maintained throughout the development of dysplasia and carcinogenesis, but more controlled studies are needed to define its role in fibrosis [54].

3.2. MicroRNAs are associated with fibrosis and strictures in CD

Transmural inflammation is a hallmark of CD leading to irreversible fibrosis and stricture formation that marks disease severity. Several studies have attempted to identify the role of microRNAs in CD-related fibrosis. MicroRNA profiling of the serum from patients with CD with and without strictures has implicated miR-19a-3p and miR-19b-3p as potential pathogenic markers [55]. The authors found that low levels of both miR-19a-3p and miR-19b-3p were strongly correlated with stricturing CD and were independent of other potentially confounding variables such as site, gender, age, disease duration, and activity [55]. The studies in liver fibrosis have implemented miR-19b as a negative regulator of the pro-fibrotic tumor growth factor-β (TGF)-β-signaling pathway, but these experiments have not been confirmed in the setting of CD-associated fibrosis [56].

Several other microRNAs have been associated with TGF-β-signaling pathway and fibrosis in CD. TGF-β signaling has been shown to promote epithelial-mesenchymal transition, which leads to fibrosis in certain contexts. In patients with stricturing CD, the level of miR-29b was assessed in the mucosa overlying a stricture by comparing mucosa-overlying areas not affected by fibrosis [57]. Furthermore, the overexpression of miR-29b in fibroblast caused a decrease in TGF-β-mediated collagen deposition [57].

The complex interactions in the microenvironment between the mucosa and underlying stroma in CD are highlighted by studies of miR-200b. In IBD, the ability of the mucosa to withstand damage and remain intact is a crucial mechanism to limit disease severity. Researchers found that the miR-200b level was decreased in the mucosa of UC and CD, which correlated with the extent of damage incurred by the epithelium [58, 59]. MiR-200b has been shown to inhibit TGF-β-mediated epithelial-mesenchymal transition through the targeting of ZEB1 and SMAD2 expression [59]. Additionally, the overexpression of miR-200b promoted the growth of epithelial cells by stimulating cyclin D1 production [59]. These studies support that miR-200b can protect against damage from CD in both epithelium and underlying stroma.

These recent observations are laying the foundation for how microRNAs are intricately involved in the formation of fibrosis and strictures in idiopathic IBD.

Advertisement

4. The role of microRNAs in IBD-associated carcinogenesis

Molecular mechanisms of IBD-associated carcinogenesis are poorly understood and are an exciting area of research within the field [60, 61]. Multiple epidemiologic and basic science studies have shown that the risk of IBD-associated colon cancer increases as the extent of the disease, severity of inflammation, and duration increase [6264]. Colonoscopies with multiple spatially distinct biopsies are used to assess for IBD-associated dysplasia. Although histologic examination can reproducibly identify dysplasia, IBD-associated dysplasia is difficult to be distinguished from sporadic adenoma based on histologic appearance alone, but certain features and patient characteristics can be helpful in suggesting IBD-associated carcinoma [65].

Molecular alterations have been shown to lead to dysplasia and carcinogenesis and the abnormalities have been demonstrated in normal-appearing nondysplastic mucosa from UC patients who had a remote dysplastic lesion [6671]. Using microRNA expression microarrays from tissue with IBD-associated dysplasia, the dysplastic epithelium revealed that 22 microRNAs (miR-31, miR-31*, miR-96, miR-135b, miR-141, miR-183, miR-192, miR-192*, miR-194, miR-194*, miR-200a, miR-200a*, miR-200b, miR-200b*, miR-200c, miR-203, miR-215, miR-224, miR-375, miR-424*, miR-429, and miR-552) were upregulated. Ten microRNAs (miR-122, miR-139-5p, miR-142-3p, miR-146b-5p, miR-155, miR-223, miR-490-2p, miR-501-5p, miR-892b, and miR-1288) have been found to be downregulated [72].

Studies of microRNAs may elucidate distinct pathways to identify IBD-associated dysplasia and subsequent carcinogenesis from sporadic mutations. As such, developing reliable molecular markers to distinguish sporadic adenoma from IBD-associated carcinogenesis will aid in the surgical management of IBD patients who are under consideration for a total colectomy.

4.1. MiR-31 as an emerging biomarker of IBD-associated dysplasia

An assessment of the different anatomic locations in normal colon showed an equally low baseline expression of miR-31 [72]. MiR-31 is upregulated in both UC and CD, but not in other inflammatory conditions that have no association with dysplasia or malignancy [73]. Subsequently, the level of miR-31 was found to be 11-fold higher in IBD-associated dysplasia and carcinoma compared to that of IBD tissue without dysplasia, although no difference was seen in miR-31 expression level between IBD-associated dysplasia and IBD-associated carcinomas [72]. However, miR-31 was also found to be upregulated in sporadic colorectal adenocarcinomas [7476].

The role of miR-31 in IBD-associated dysplasia or malignancy has only recently been examined and it is too early to determine if it is a useful marker of IBD-associated dysplasia or a generalized biomarker for carcinoma.

4.2. MiR-214 as an emerging oncogenic driver of IBD-associated carcinogenesis

Recent evidence suggests that microRNAs are not just biomarkers, as they can be crucial for carcinogenesis. A high-throughput genomic screen of microRNAs has identified miR-214 as a positive regulator of NF-kB activity [77]. Elevated levels of miR-214 were found in human tissue from patients with active UC or CD [77]. Two groups have previously shown that miR-214 promotes the growth of malignant osteosarcomas by directly reducing levels of the tumor suppressor, phosphatase and tensin homology [78, 79]. Using in vitro and in vivo models, it was displayed that chronic disease activity initiated interleukin-6 causing STAT3-mediated transcription of miR-214 with subsequent reduction of PTEN, PDZ, and LIM domain 2 (PDLIM2) causing enhanced activity of oncogenic NF-kB [77]. Furthermore, an inhibitor of miR-214 was able to inhibit DSS-induced carcinogenesis [77]. These data support not only the role of miR-214 as an oncogene but also provide insight into the molecular mechanisms of its carcinogenesis.

4.3. MiR-124a as a tumor suppressor epigenetically inactivated in IBD-associated carcinogenesis

Chronic inflammation has been shown to cause hypermethylation of CpG islands within the promoter of genes causing decreased gene expression [80]. Epigenetic silencing of well-characterized tumor suppressor proteins, E-cadherin, p14, and hMLH1, has been documented in UC-associated carcinogenesis [8184].

The role of miR-124a as a tumor suppressor has been shown by demonstrating the direct suppression of oncogenic cyclin-dependent kinase 6 (CDK6) in acute lymphoblastic leukemia, medulloblastoma, and hepatocellular carcinoma [8587]. The evaluation of patients with UC showed a strong correlation between increased miR-124a-3 hypermethylation and increased CDK6 expression [88]. The levels of methylation were highest in patients who had pancolitis and long-standing disease, and the individual with the highest value was later found to have high-grade dysplasia [88]. These studies are supported by the usage of high-throughput genetic screens that identified miR-124a as a negative regulator of oncogenic STAT3 in human colonic epithelial cells [89]. The authors demonstrated that miR-124a was downregulated through hypermethylation in pediatric patients with UC and strongly correlated with elevated levels of STAT3 and increased disease severity [89].

In combination, these studies demonstrate that the long-standing inflammation of IBD has the power to epigenetically downregulate important microRNA tumor suppressors that alters the downstream targets of those events. These findings might open up novel pathways that can be assessed for prognosis and potential personalized medicine.

Advertisement

5. MicroRNA as potential therapeutic targets for IBD

The unique ability of microRNAs to posttranscriptionally regulate gene expression and affect multiple biological signaling pathways can lead to the development of antisense oligonucleotides as potential novel therapeutics in IBD. Multiple studies have shown that antisense oligonucleotides complementary to microRNAs can target specific microRNAs, abolishing their function in both in vitro cell models and in vivo animal models. Preclinical cell- and animal-based models have demonstrated that altering microRNA levels can modify the expression of either tumor suppressors or oncogenes as such to effect cancer growth [9093]. Additionally, drug-induced microRNA changes have been shown to contribute to the therapeutic effect in in vitro models [94, 95].

With specific regards to the field of IBD, potential therapeutic targets consist of three general strategies: (1) to provide antisense oligonucleotides that inactivate microRNAs that are pro-inflammatory; (2) to replace the expression of tumor suppressor microRNAs; and (3) to provide antisense oligonucleotides that inactivate oncogenic microRNAs.

At the time of publication, no therapeutic manipulation of microRNAs in IBD has been published in either cell lines or animal models. However, a recent study has shown that the inhibition of miR-21, a central driver of IBD pathogenesis, slows the proliferation of a nasopharyngeal carcinoma cell line [96].

Although the delivery of microRNA-targeted therapeutics is technically challenging, recent advances in delivery methods suggest that we may soon see an explosion of microRNA-targeted therapies being assessed in vivo for the first time [9799].

Advertisement

6. Conclusions

In summary, the rapidly expanding knowledge of microRNAs in the pathogenesis and carcinogenesis associated with IBD has created an emerging interest in their potential role for personalized therapies.

References

  1. 1. Xavier, R.J. and Podolsky, D.K., Unravelling the pathogenesis of inflammatory bowel disease. Nature, 2007. 448(7152): p. 427–34.
  2. 2. Kugathasan, S. and Amre, D., Inflammatory bowel disease—environmental modification and genetic determinants. Pediatr Clin North Am, 2006. 53(4): p. 727–49.
  3. 3. Maloy, K.J. and Powrie, F., Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature, 2011. 474(7351): p. 298–306.
  4. 4. Kozomara, A. and Griffiths-Jones, S., miRBase: integrating microRNA annotation and deep-sequencing data. Nucleic Acids Res, 2011. 39(Database issue): p. D152–7.
  5. 5. Kozomara, A. and Griffiths-Jones, S., miRBase: annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res, 2014. 42(Database issue): p. D68–73.
  6. 6. Ambros, V., The functions of animal microRNAs. Nature, 2004. 431(7006): p. 350–5.
  7. 7. Bartel, D.P., MicroRNAs: target recognition and regulatory functions. Cell, 2009. 136(2): p. 215–33.
  8. 8. Saini, H.K., Enright, A.J., and Griffiths-Jones, S., Annotation of mammalian primary microRNAs. BMC Genomics, 2008. 9: p. 564.
  9. 9. Filipowicz, W., Bhattacharyya, S.N., and Sonenberg, N., Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat Rev Genet, 2008. 9(2): p. 102–14.
  10. 10. Calin, G.A. and Croce, C.M., MicroRNA signatures in human cancers. Nat Rev Cancer, 2006. 6(11): p. 857–66.
  11. 11. Esteller, M., Non-coding RNAs in human disease. Nat Rev Genet, 2011. 12(12): p. 861–74.
  12. 12. Kerr, T.A., Korenblat, K.M., and Davidson, N.O., MicroRNAs and liver disease. Transl Res, 2011. 157(4): p. 241–52.
  13. 13. Kloosterman, W.P. and Plasterk, R.H., The diverse functions of microRNAs in animal development and disease. Dev Cell, 2006. 11(4): p. 441–50.
  14. 14. Lorenzen, J.M., Haller, H., and Thum, T., MicroRNAs as mediators and therapeutic targets in chronic kidney disease. Nat Rev Nephrol, 2011. 7(5): p. 286–94.
  15. 15. Lu, J., et al., MicroRNA expression profiles classify human cancers. Nature, 2005. 435(7043): p. 834–8.
  16. 16. Wu, F., et al., MicroRNAs are differentially expressed in ulcerative colitis and alter expression of macrophage inflammatory peptide-2 alpha. Gastroenterology, 2008. 135(5): p. 1624–1635 e24.
  17. 17. Takagi, T., et al., Increased expression of microRNA in the inflamed colonic mucosa of patients with active ulcerative colitis. J Gastroenterol Hepatol, 2010. 25(Suppl 1): p. S129–33.
  18. 18. Feng, X., et al., Up-regulation of microRNA-126 may contribute to pathogenesis of ulcerative colitis via regulating NF-kappaB inhibitor IkBα. PLoS One, 2012. 7(12): p. e52782.
  19. 19. Fasseu, M., et al., Identification of restricted subsets of mature microRNA abnormally expressed in inactive colonic mucosa of patients with inflammatory bowel disease. PLoS One, 2010. 5(10): p. e13160.
  20. 20. Bian, Z., et al., Role of miR-150-targeting c-Myb in colonic epithelial disruption during dextran sulphate sodium-induced murine experimental colitis and human ulcerative colitis. J Pathol, 2011. 225(4): p. 544–53.
  21. 21. Pekow, J.R., et al., miR-143 and miR-145 are downregulated in ulcerative colitis: putative regulators of inflammation and protooncogenes. Inflamm Bowel Dis, 2012. 18(1): p. 94–100.
  22. 22. Coskun, M., et al., miR-20b, miR-98, miR-125b-1*, and let-7e* as new potential diagnostic biomarkers in ulcerative colitis. World J Gastroenterol, 2013. 19(27): p. 4289–99.
  23. 23. Beres, N.J., et al., Role of altered expression of miR-146a, miR-155, and miR-122 in pediatric patients with inflammatory bowel disease. Inflamm Bowel Dis, 2016. 22(2): p. 327–35.
  24. 24. Lin, J., et al., Novel specific microRNA biomarkers in idiopathic inflammatory bowel disease unrelated to disease activity. Mod Pathol, 2014. 27(4): p. 602–8.
  25. 25. Koukos, G., et al., A microRNA signature in pediatric ulcerative colitis: deregulation of the miR-4284/CXCL5 pathway in the intestinal epithelium. Inflamm Bowel Dis, 2015. 21(5): p. 996–1005.
  26. 26. Yang, Y., et al., Overexpression of miR-21 in patients with ulcerative colitis impairs intestinal epithelial barrier function through targeting the Rho GTPase RhoB. Biochem Biophys Res Commun, 2013. 434(4): p. 746–52.
  27. 27. Zahm, A.M., et al., Rectal microRNAs are perturbed in pediatric inflammatory bowel disease of the colon. J Crohns Colitis, 2014. 8(9): p. 1108–17.
  28. 28. Iborra, M., et al., Identification of serum and tissue micro-RNA expression profiles in different stages of inflammatory bowel disease. Clin Exp Immunol, 2013. 173(2): p. 250–8.
  29. 29. Wu, F., et al., Identification of microRNAs associated with ileal and colonic Crohn's disease. Inflamm Bowel Dis, 2010. 16(10): p. 1729–38.
  30. 30. Nguyen, H.T., et al., MicroRNA-7 modulates CD98 expression during intestinal epithelial cell differentiation. J Biol Chem, 2010. 285(2): p. 1479–89.
  31. 31. Brest, P., et al., A synonymous variant in IRGM alters a binding site for miR-196 and causes deregulation of IRGM-dependent xenophagy in Crohn's disease. Nat Genet, 2011. 43(3): p. 242–5.
  32. 32. Huang, Y., et al., miR-141 regulates TGF-beta1-induced epithelial-mesenchymal transition through repression of HIPK2 expression in renal tubular epithelial cells. Int J Mol Med, 2015. 35(2): p. 311–8.
  33. 33. Nguyen, H.T., et al., Crohn's disease-associated adherent invasive Escherichia coli modulate levels of microRNAs in intestinal epithelial cells to reduce autophagy. Gastroenterology, 2014. 146(2): p. 508–19.
  34. 34. Wu, F., et al., Peripheral blood microRNAs distinguish active ulcerative colitis and Crohn's disease. Inflamm Bowel Dis, 2011. 17(1): p. 241–50.
  35. 35. Paraskevi, A., et al., Circulating MicroRNA in inflammatory bowel disease. J Crohns Colitis, 2012. 6(9): p. 900–4.
  36. 36. Krissansen, G.W., et al., Overexpression of miR-595 and miR-1246 in the sera of patients with active forms of inflammatory bowel disease. Inflamm Bowel Dis, 2015. 21(3): p. 520–30.
  37. 37. Polytarchou, C., et al., Assessment of circulating microRNAs for the diagnosis and disease activity evaluation in patients with ulcerative colitis by using the nanostring technology. Inflamm Bowel Dis, 2015. 21(11): p. 2533–9.
  38. 38. Zahm, A.M., et al., Circulating microRNA is a biomarker of pediatric Crohn disease. J Pediatr Gastroenterol Nutr, 2011. 53(1): p. 26–33.
  39. 39. Duttagupta, R., et al., Genome-wide maps of circulating miRNA biomarkers for ulcerative colitis. PLoS One, 2012. 7(2): p. e31241.
  40. 40. Meucci, G., et al., Frequency and clinical evolution of indeterminate colitis: a retrospective multi-centre study in northern Italy. GSMII (Gruppo di Studio per le Malattie Infiammatorie Intestinali). Eur J Gastroenterol Hepatol, 1999. 11(8): p. 909–13.
  41. 41. Moum, B., et al., Incidence of ulcerative colitis and indeterminate colitis in four counties of southeastern Norway, 1990-93. A prospective population-based study. The Inflammatory Bowel South-Eastern Norway (IBSEN) Study Group of Gastroenterologists. Scand J Gastroenterol, 1996. 31(4): p. 362–6.
  42. 42. Lin, J., et al., MicroRNA expression patterns in indeterminate inflammatory bowel disease. Mod Pathol, 2013. 26(1): p. 148–154.
  43. 43. Adams, A.T., et al., Two-stage genome-wide methylation profiling in childhood-onset Crohn's disease implicates epigenetic alterations at the VMP1/MIR21 and HLA loci. Inflamm Bowel Dis, 2014. 20(10): p. 1784–93.
  44. 44. Han, L., et al., DNA methylation regulates microRNA expression. Cancer Biol Ther, 2007. 6(8): p. 1284–8.
  45. 45. Shi, C., et al., MicroRNA-21 knockout improve the survival rate in DSS induced fatal colitis through protecting against inflammation and tissue injury. PLoS One, 2013. 8(6): p. e66814.
  46. 46. Lv, C., et al., Role and mechanism of microRNA-21 in HO-induced apoptosis in bone marrow mesenchymal stem cells. J Clin Neurosci, 2016, 27: p. 154–60.
  47. 47. Zhang, W., W. Bai, and W. Zhang, MiR-21 suppresses the anticancer activities of curcumin by targeting PTEN gene in human non-small cell lung cancer A549 cells. Clin Transl Oncol, 2014. 16(8): p. 708–13.
  48. 48. Yu, X., et al., Silencing of MicroRNA-21 confers the sensitivity to tamoxifen and fulvestrant by enhancing autophagic cell death through inhibition of the PI3K-AKT-mTOR pathway in breast cancer cells. Biomed Pharmacother, 2016. 77: p. 37–44.
  49. 49. Lv, C., Hao, Y., and Tu, G., MicroRNA-21 promotes proliferation, invasion and suppresses apoptosis in human osteosarcoma line MG63 through PTEN/Akt pathway. Tumour Biol, 2016, Jan 16, DOI: 10.1007/s13277-016-4807-6.
  50. 50. Liu, X., et al., MiR-21 inhibits autophagy by targeting Rab11a in renal ischemia/reperfusion. Exp Cell Res, 2015. 338(1): p. 64–9.
  51. 51. Zarjou, A., et al., Identification of a microRNA signature in renal fibrosis: role of miR-21. Am J Physiol Renal Physiol, 2011. 301(4): p. F793–801.
  52. 52. Yang, G., et al., Discovery and validation of extracellular/circulating microRNAs during idiopathic pulmonary fibrosis disease progression. Gene, 2015. 562(1): p. 138–44.
  53. 53. Zhao, J., et al., MiR-21 simultaneously regulates ERK1 signaling in HSC activation and hepatocyte EMT in hepatic fibrosis. PLoS One, 2014. 9(10): p. e108005.
  54. 54. Ludwig, K., et al., PDCD4/miR-21 dysregulation in inflammatory bowel disease-associated carcinogenesis. Virchows Arch, 2013. 462(1): p. 57–63.
  55. 55. Lewis, A., et al., Low serum levels of microRNA-19 are associated with a stricturing Crohn's disease phenotype. Inflamm Bowel Dis, 2015. 21(8): p. 1926–34.
  56. 56. Lakner, A.M., et al., Inhibitory effects of microRNA 19b in hepatic stellate cell-mediated fibrogenesis. Hepatology, 2012. 56(1): p. 300–10.
  57. 57. Nijhuis, A., et al., In Crohn's disease fibrosis-reduced expression of the miR-29 family enhances collagen expression in intestinal fibroblasts. Clin Sci (Lond), 2014. 127(5): p. 341–50.
  58. 58. Chen, Y., et al., miR-200b is involved in intestinal fibrosis of Crohn's disease. Int J Mol Med, 2012. 29(4): p. 601–6.
  59. 59. Chen, Y., et al., miR-200b inhibits TGF-beta1-induced epithelial-mesenchymal transition and promotes growth of intestinal epithelial cells. Cell Death Dis, 2013. 4: p. e541.
  60. 60. Eaden, J.A., Abrams, K.R., and Mayberry, J.F., The risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut, 2001. 48(4): p. 526–35.
  61. 61. Jess, T., et al., Increased risk of intestinal cancer in Crohn's disease: a meta-analysis of population-based cohort studies. Am J Gastroenterol, 2005. 100(12): p. 2724–9.
  62. 62. Rutter, M., et al., Severity of inflammation is a risk factor for colorectal neoplasia in ulcerative colitis. Gastroenterology, 2004. 126(2): p. 451–9.
  63. 63. Gupta, R.B., et al., Histologic inflammation is a risk factor for progression to colorectal neoplasia in ulcerative colitis: a cohort study. Gastroenterology, 2007. 133(4): p. 1099–105; quiz 1340–1.
  64. 64. Rubin, D.T., The changing face of colorectal cancer in inflammatory bowel disease: progress at last! Gastroenterology, 2006. 130(4): p. 1350–2.
  65. 65. Liu, X., et al., Distinct clinicohistologic features of inflammatory bowel disease-associated colorectal adenocarcinoma: in comparison with sporadic microsatellite-stable and Lynch syndrome-related colorectal adenocarcinoma. Am J Surg Pathol, 2012. 36(8): p. 1228–33.
  66. 66. Brentnall, T.A., et al., Mutations in the p53 gene: an early marker of neoplastic progression in ulcerative colitis. Gastroenterology, 1994. 107(2): p. 369–78.
  67. 67. Burmer, G.C., et al., Neoplastic progression in ulcerative colitis: histology, DNA content, and loss of a p53 allele. Gastroenterology, 1992. 103(5): p. 1602–10.
  68. 68. Rubin, C.E., et al., DNA aneuploidy in colonic biopsies predicts future development of dysplasia in ulcerative colitis. Gastroenterology, 1992. 103(5): p. 1611–20.
  69. 69. Chen, R., et al., DNA fingerprinting abnormalities can distinguish ulcerative colitis patients with dysplasia and cancer from those who are dysplasia/cancer-free. Am J Pathol, 2003. 162(2): p. 665–72.
  70. 70. Rabinovitch, P.S., et al., Pancolonic chromosomal instability precedes dysplasia and cancer in ulcerative colitis. Cancer Res, 1999. 59(20): p. 5148–53.
  71. 71. Okahara, S., et al., Inflammatory gene signature in ulcerative colitis with cDNA macroarray analysis. Aliment Pharmacol Ther, 2005. 21(9): p. 1091–7.
  72. 72. Olaru, A.V., et al., Dynamic changes in the expression of MicroRNA-31 during inflammatory bowel disease-associated neoplastic transformation. Inflamm Bowel Dis, 2011. 17(1): p. 221–31.
  73. 73. Zhang, C., et al., Differential expression of miR-31 between inflammatory bowel disease and microscopic colitis. MicroRNA, 2014. 3(3): p. 155–9.
  74. 74. Xu, X.M., et al., Expression of miR-21, miR-31, miR-96 and miR-135b is correlated with the clinical parameters of colorectal cancer. Oncol Lett, 2012. 4(2): p. 339–45.
  75. 75. Slaby, O., et al., Altered expression of miR-21, miR-31, miR-143 and miR-145 is related to clinicopathologic features of colorectal cancer. Oncology, 2007. 72(5-6): p. 397–402.
  76. 76. Schee, K., et al., Clinical relevance of microRNA miR-21, miR-31, miR-92a, miR-101, miR-106a and miR-145 in colorectal cancer. BMC Cancer, 2012. 12: p. 505.
  77. 77. Polytarchou, C., et al., MicroRNA214 is associated with progression of ulcerative colitis, and inhibition reduces development of colitis and colitis-associated cancer in mice. Gastroenterology, 2015. 149(4): p. 981–992.e11.
  78. 78. Liu, C.J., et al., MiR214 promotes osteosarcoma tumor growth and metastasis by decreasing the expression of PTEN. Mol Med Rep, 2015. 12(4): p. 6261–6.
  79. 79. Wang, X., et al., MicroRNA-214 regulates osteosarcoma survival and growth by directly targeting phosphatase and tensin homolog. Mol Med Rep, 2014. 10(6): p. 3073–9.
  80. 80. Niwa, T. and Ushijima, T., Induction of epigenetic alterations by chronic inflammation and its significance on carcinogenesis. Adv Genet, 2010. 71: p. 41–56.
  81. 81. Azarschab, P., et al., Epigenetic control of the E-cadherin gene (CDH1) by CpG methylation in colectomy samples of patients with ulcerative colitis. Genes Chromosomes Cancer, 2002. 35(2): p. 121–6.
  82. 82. Wheeler, J.M., et al., Hypermethylation of the promoter region of the E-cadherin gene (CDH1) in sporadic and ulcerative colitis associated colorectal cancer. Gut, 2001. 48(3): p. 367–71.
  83. 83. Fleisher, A.S., et al., Microsatellite instability in inflammatory bowel disease-associated neoplastic lesions is associated with hypermethylation and diminished expression of the DNA mismatch repair gene, hMLH1. Cancer Res, 2000. 60(17): p. 4864–8.
  84. 84. Moriyama, T., et al., Hypermethylation of p14 (ARF) may be predictive of colitic cancer in patients with ulcerative colitis. Dis Colon Rectum, 2007. 50(9): p. 1384–92.
  85. 85. Agirre, X., et al., Epigenetic silencing of the tumor suppressor microRNA Hsa-miR-124a regulates CDK6 expression and confers a poor prognosis in acute lymphoblastic leukemia. Cancer Res, 2009. 69(10): p. 4443–53.
  86. 86. Pierson, J., et al., Regulation of cyclin dependent kinase 6 by microRNA 124 in medulloblastoma. J Neurooncol, 2008. 90(1): p. 1–7.
  87. 87. Furuta, M., et al., miR-124 and miR-203 are epigenetically silenced tumor-suppressive microRNAs in hepatocellular carcinoma. Carcinogenesis, 2010. 31(5): p. 766–76.
  88. 88. Ueda, Y., et al., DNA methylation of microRNA-124a is a potential risk marker of colitis-associated cancer in patients with ulcerative colitis. Dig Dis Sci, 2014. 59(10): p. 2444–51.
  89. 89. Koukos, G., et al., MicroRNA-124 regulates STAT3 expression and is down-regulated in colon tissues of pediatric patients with ulcerative colitis. Gastroenterology, 2013. 145(4): p. 842–52.e2.
  90. 90. Krutzfeldt, J., et al., Silencing of microRNAs in vivo with 'antagomirs'. Nature, 2005. 438(7068): p. 685–9.
  91. 91. Meister, G. and Tuschl, T., Mechanisms of gene silencing by double-stranded RNA. Nature, 2004. 431(7006): p. 343–9.
  92. 92. Elmen, J., et al., LNA-mediated microRNA silencing in non-human primates. Nature, 2008. 452(7189): p. 896–9.
  93. 93. Tong, A.W. and Nemunaitis, J., Modulation of miRNA activity in human cancer: a new paradigm for cancer gene therapy? Cancer Gene Ther, 2008. 15(6): p. 341–55.
  94. 94. Rossi, L., Bonmassar, E., and Faraoni, I., Modification of miR gene expression pattern in human colon cancer cells following exposure to 5-fluorouracil in vitro. Pharmacol Res, 2007. 56(3): p. 248–53.
  95. 95. Blower, P.E., et al., MicroRNA expression profiles for the NCI-60 cancer cell panel. Mol Cancer Ther, 2007. 6(5): p. 1483–91.
  96. 96. Li, Y., et al., miR-21 inhibitor suppresses proliferation and migration of nasopharyngeal carcinoma cells through down-regulation of BCL2 expression. Int J Clin Exp Pathol, 2014. 7(6): p. 3478–87.
  97. 97. Wang, H., et al., Recent progress in microRNA delivery for cancer therapy by non-viral synthetic vectors. Adv Drug Deliv Rev, 2015. 81: p. 142–60.
  98. 98. Ben-Shushan, D., et al., Overcoming obstacles in microRNA delivery towards improved cancer therapy. Drug Deliv Transl Res, 2014. 4(1): p. 38–49.
  99. 99. Esposito, C.L., Catuogno, S., and de Franciscis, V., Aptamer-MiRNA conjugates for cancer cell-targeted delivery. Methods Mol Biol, 2016. 1364: p. 197–208.

Written By

Kurt Fisher and Jingmei Lin

Submitted: 20 October 2015 Reviewed: 07 July 2016 Published: 26 October 2016