Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Genomics Proteomics Bioinformatics. 2012 Oct; 10(5): 246–253.
Published online 2012 Sep 29. doi: 10.1016/j.gpb.2012.07.005
PMCID: PMC3611977
NIHMSID: NIHMS453312
PMID: 23200134

MicroRNAs in Common Human Diseases

Abstract

MicroRNAs (miRNAs) are a class of short non-coding RNA molecules that have attracted tremendous attention from the biological and biomedical research communities over the past decade. With over 1900 miRNAs discovered in humans to date, many of them have already been implicated in common human disorders. Facilitated by high-throughput genomics and bioinformatics in conjunction with traditional molecular biology techniques and animal models, miRNA research is now positioned to make the transition from laboratories to clinics to deliver profound benefits to public health. Herein, we overview the progress of miRNA research related to human diseases, as well as the potential for miRNA to becoming the next generation of diagnostics and therapeutics.

Keywords: MicroRNA, Human diseases, Diagnostics, Therapeutics, Biomarker

A historical overview of microRNA research

MicroRNAs (miRNAs) are a class of recently identified non-coding RNA molecules that play an essential role in gene expression regulation at post-transcriptional levels [1]. With the first miRNA, lin-4, discovered in Caenorhabditis elegans in 1993 via forward genetics [2], the second C. elegans miRNA, let-7, was not identified by the same approach until seven years later [3]. This time gap highlights not only the inefficiency of forward genetics and standard molecular biology techniques to discover miRNAs, but also the lack of enthusiasm among researchers who previously suspected that miRNA was merely a worm-specific phenomenon. However, the field of miRNA research has since flourished with over 17,000 miRNAs discovered to date in 142 species, including more than 1900 in humans [4]. The key word “miRNA” currently pulls more than 16,000 publications from PubMed, and the first miRNA-targeted drug has now entered a phase II clinical trial (http://www.ClinicalTrials.gov), demonstrating early promise. In retrospect, the timing of miRNA research evolution was particularly interesting as it echoed the time frame of the Human Genome Project (HGP) and many other whole-genome sequencing projects completed over the past decade. The completion of these projects has impacted the field of miRNA research in profound ways.

The fruitful expansion of miRNA research was triggered by the identification and functional characterization of let-7 [3]. When Ruvkun et al. demonstrated that the let-7 sequence was highly conserved across the evolutionary spectrum [5], biologists started to realize that this tiny RNA molecule may have a big role to play in humans as well [6]. Before long, three competing laboratories made de novo identifications of dozens of single-stranded RNA molecules approximately 22 nt in length by the combination of an improved cloning method and bioinformatics, a novel approach at the time [7], [8], [9]. The method of de novo identification was rather successful, leading to most miRNA discoveries before 2006, including more than 300 in humans. More importantly, it revealed the intrinsic characteristics of miRNA as a class, such as the secondary structure of miRNA precursors, allowing new miRNAs to be computationally identified. However, the de novo identification method came with a few limitations. It was difficult to clone miRNAs expressed at low levels or with certain sequence compositions and post-transcriptional modifications [10], [11]. Nevertheless, these limitations could be bypassed through in silico prediction. With the completion of many whole genome sequencing projects [12], [13], [14], [15], thousands of new miRNA species were now identifiable by computational prediction [4], [16]. Taking a variety of factors into consideration, such as sequence conservation and thermodynamic stability of secondary structure, researchers were now able to identify new miRNA species that failed to be discovered by cloning approaches [17]. To date, the vast majority of known miRNA species have been discovered by bioinformatics and their sequences can be found in the Sanger miRNA registry (http://www.miRBase.com), an open access database for miRNA research [4].

The intriguing story of miRNA cannot be fully revealed without identifying miRNA targets in the context of biological processes. Through painstaking characterization of miRNA biogenesis and functional pathways [18], it is now clear that miRNAs repress the expression of cellular gene targets in a sequence-dependent manner. Specifically, the miRNA “seed”, i.e., the sequence between the 2nd and the 8th nt from the 5′ end, is essential in recognizing targets [19]. Facilitated by Dicer, an RNase III family member, the heteroduplex of miRNA and its target mRNA is integrated into the RNA-induced silencing complex (RISC). Mainly composed with the multi-functional catalytic protein, Argonaute, and a double stranded RNA binding protein, TRBP, responsible for recruiting Dicer to Argonaute [20], [21], [22], [23], RISC plays a central role in miRNA-mediated repression on gene expression [21]. The type of repression relies on the degree of sequence complementarity between seed and target sequences. Whereas partial complementarity may induce translation repression or target mRNA instability, perfect complementarity normally causes target mRNA destruction [24]. This target recognition mechanism allows for in silico methods of target prediction by aligning miRNA sequences with entire genomes in searching for potential miRNA binding sites. Adopting similar algorithms, a few groups have developed open access target prediction software with minor variations, such as miRBase, PicTar, TargetScan and miRanda [25], [26], [27], [28], [29], [30], etc. Conversely, researchers have been trying to use high-throughput genomic approaches, such as oligonucleotide microarrays facilitated by bioinformatics, to experimentally identify targets [31], [32], [33]. Evidence suggests that RNA destabilization is the predominant mechanism mediated by miRNA in mammals, making these methods particularly useful for identifying strong miRNA complementarities with marked effects [34]. Specifically, by introducing a miRNA of interest into cultured mammalian cells, the expression changes of predicted targets are monitored in real-time [31]. Sequence alignment of the artificially over-expressed miRNA and the down-regulated mRNA would further suggest a direct regulation or off-target effect. Both in silico prediction and target expression profiling suggest that the regulatory relationships of miRNAs and their targets are complex. Because of short seed sequences, multiple miRNAs may repress the expression of a specific gene simultaneously by targeting different sequence regions; likewise, a single miRNA may be able to regulate the expression of dozens or even hundreds of targets at the same time. Although it was initially believed that miRNA-mediated repression takes place exclusively in the cytoplasm, new evidence suggests that it may also occur in other cellular compartments such as mitochondria and nucleus [35], [36]. The complexity of regulation underscores the necessity of combining traditional molecular biology with modern bioinformatic approaches to characterize the roles of miRNA more effectively.

miRNAs and human diseases

As discovery of human miRNAs increased, the research focus was gradually shifted towards functional characterization of miRNAs, particularly in the context of human diseases. The connection between miRNAs and disease was obvious. miRNA expression patterns are tissue-specific [37] and in many cases define the physiological nature of the cell [31]. The definitive evidence came from a report demonstrating that the gene expression profile of a non-neuron cell became more like that of a neuron when the neuron-specific miR-124 was artificially over-expressed within [31]. If the same premise holds true, certain miRNA expression patterns could be disease-specific and hold great prognostic value. In fact, a more comprehensive miRNA profiling study demonstrated that distinct miRNA expression patterns were specific to various types of cancers and were able to reflect the developmental lineage and differentiation state of tumors [38]. More specifically, many miRNAs were found to play key roles in vital biological processes such as cell division and death [39], cellular metabolism [40], intracellular signaling [41], immunity [42] and cell movement [43]. Therefore, aberrant miRNA expression should proportionately affect those critical processes, and as a result, lead to various pathological and occasionally malignant outcomes. Here, we overview miRNA-related studies focused on high-priority human diseases with insufficient treatment options (Table 1).

Table 1

miRNAs associated with common human diseases

DISEASEMIRNAREFERENCE
CancerB-CLLmiR-15, miR-16[44]
Breast cancermiR-125b, miR-145, miR-21, miR-155, miR-210[46], [56]
Lung cancermiR-155, let-7a[47]
Gastric cancermiR-145[54]
Liver cancermiR-29b[57], [58]


Viral diseasesHCVmiR-122, miR-155[72], [73], [78]
HIV-1miR-28, miR-125b, miR-150, miR-223, miR-382[75]
Influenza virusmiR-21, miR-223[76], [77]


Immune-related diseasesMultiple sclerosismiR-145, miR-34a, miR-155, miR-326[80], [81]
Systemic lupus erythematosusmiR-146a[82], [83]
Type II diabetesmiR-144, miR-146a, miR-150, miR-182, miR-103, miR-107[84]
Nonalcoholic fatty liver diseasemiR-200a, miR-200b, miR-429, miR-122, miR-451, miR-27[86]
Non-alcoholic steatohepatitismiR-29c, miR-34a, miR-155, miR-200b[87]


Neurodegenerative diseasesParkinson’s diseasemiR-30b, miR-30c, miR-26a, miR-133b, miR-184, let-7[90], [91], [92]
Alzheimer’s diseasemiR-29b-1, miR-29a, miR-9[94]

Cancers

Since the early stages of miRNA research, cancer has been the most prominent of human diseases with a clear role for miRNA regulation. The first evidence came from a study by Calin et al. in which they demonstrated a frequent deletion of miRNA genes miR15 and miR16 among 65% of B-cell chronic lymphocytic leukemia (B-CLL) patients [44]. Intriguingly, down-regulation of miR-15 and miR-16 expression was observed among B-CLL patients without the deletion, suggesting that the pathogenesis of B-CLL may be attributed to the intracellular abundance of two miRNAs. Encouraged by this finding, this group applied a systemic search on the complete human genome and established correlations of miRNAs with various cancers [45]. Subsequent expression profiling studies further demonstrated the correlation between aberrant miRNA expression patterns and increased occurrence of different types of cancers. Notably, the deregulation of miR-125b, miR-145, miR-21, and miR-155 expression was associated with the increased risk of breast cancer [46]. In addition, up-regulation of miR-155 and down-regulation of let-7a were correlated with poor survival of lung cancer patients [47], indicating an imbalance of cell death and proliferation during cancer development [48], [49], [50]. Intriguingly, miRNA expression patterns were also able to stage cancer progression [38], indicating that miRNA levels were not only useful in diagnosis but also potentially in prognosis of diseases. These cancer-related miRNAs were categorized into tumor suppressors and oncogenes due to their associations with opposite clinical outcomes with altered expressions. For example, miR-15, miR-16 and let-7 are known tumor suppressors while miR-21 and miR-155 serve as oncogenes [44], [51], [52].

The discovery of cancer-related miRNAs by expression profiling inspired mechanistic studies to implicate specific miRNAs in tumorigenesis pathways. miR-15 and miR-16 were found to repress the expression of anti-apoptotic gene bcl-2 thereby promoting cell death in cancerous cells [52]. Likewise, let-7 family members demonstrate anti-cancer properties due to their ability to repress the expression of the oncogene, ras [53]. In contrast, miR-21 directly serves as an anti-apoptotic factor in glioblastomas and breast cancer [46], [51]. Similarly, miR-155 interferes with the process of mismatch repair by repressing the expression of the MSH gene family members in colorectal cancer [54].

miRNAs also play key roles in tumor invasion and metastasis. miRNA expression profiling revealed the stepwise down-regulation of miR-145 levels with progression of primary gastric cancers and secondary metastases [55], as well as metastatic prostate cancer [56]. Similarly, increased expression of miR-210 was observed during the invasive transition of breast cancer [57]. While profiling studies establish disease correlations, mechanistic studies characterize the role of miRNAs in greater detail. For example, through the use of synthetic miRNA mimics, miR-7 and miR-29b were shown to suppress the metastasis of liver cancer by targeting PIK3CD [58] and MMP-2 [59], respectively. These cancer-related miRNAs are potentially useful for developing not only early diagnosis, but also novel anti-cancer strategies.

Viral diseases

Viruses are a group of pathogens with members causing not only severe, chronic diseases, but also some of the most deadly pandemics in human history. While miRNAs were being identified in eukaryotes, viral-encoded miRNAs were discovered in multiple virus species as well. The first viral-encoded miRNAs were cloned from a Burkitt’s lymphoma cell line latently infected by Epstein–Barr virus (EBV), a DNA virus of the herpesvirus family [60]. Soon after, dozens of viral miRNAs were identified in polyoma virus[61], adenovirus [62], and several subtypes of the herpes viruses by cloning, bioinformatics, or combined approaches [63], [64], [65]. Some preliminary evidence even suggested that RNA viruses may also encode miRNAs in spite of small genome sizes [66], [67], [68]; however, these findings have not been verified independently [65], [69].

Besides bearing viral miRNAs, alternatively, viruses are capable of regulating the expression of host cellular miRNAs for their own benefit. For example, unlike the Kaposi’s sarcoma-associated herpes virus (KHSV) which encodes a viral miRNA, miR-K12-11, EBV is able to up-regulate the expression of cellular miR-155, an ortholog of miR-K12-11 [70]. Interestingly, these two miRNAs target the same set of cellular genes, indicating a similar function [71]. A more detailed study revealed that miR-155 may prevent EBV-infected cells from apoptotic death [72], a common strategy mediated by hosts to constrain viral infection. This demonstrates the potential consequences of a virus gaining control of cellular miRNA expression for its survival.

Although the expression of some cellular miRNAs is not directly regulated by viruses, maintenance of their intracellular level is pivotal for viral infection and replication. For example, high levels of liver-specific miR-122 expression is necessary for HCV replication both in vitro and in vivo [73], [74], although viral infection and replication does not affect the expression of miR-122 [75]. On the contrary, copies of miR-28, miR-125b, miR-150, miR-223 and miR-382 are maintained at high levels in resting CD4+ T cells, but significantly decreased in activated CD4+ T cells, resulting in productive infection of HIV-1 in only the latter case [76]. These findings may help explain the tissue-specificity of virus infections and provide novel targets for anti-viral therapeutics.

Finally, miRNA expression changes may demonstrate how hosts respond to viral infections. For example, aberrant expression of a subset of cellular miRNAs was observed in lethal influenza virus infection, but not in non-lethal infection in animal models [77], [78]. Specifically, miR-21 and miR-223 were strongly up-regulated in lethal infections of H1N1 pandemic influenza virus and H5N1 avian influenza virus in mice and macaques, respectively [77], [78] while their expression was unchanged or only moderately up-regulated in animals infected with less pathogenic viruses. More recently, marked increase of miR-155 was seen in HCV-infected patients [79]. The up-regulation of miR-155 by HCV-infection may activate Wnt signaling pathway and contribute in part to HCV-induced hepatocarcinogenesis [79]. These variable miRNA expression patterns may be useful in guiding physicians to make treatment plans for patients infected by more or less virulent pathogens.

Immune-related diseases

Many common immune-related diseases, including multiple sclerosis (MS), systemic lupus erythematosus (SLE), type I/II diabetes, and nonalcoholic fatty liver disease (NAFLD), have shown established correlations with cellular miRNAs. Dozens of miRNA signatures were identified by comparing the miRNA expression profiles of relapsing-remitting MS and healthy controls [80]. Specifically, the expression of miR-145 alone was found to distinguish affected patients from healthy controls with high specificity and sensitivity. Increased expression of miR-34a, miR-155 and miR-326 was observed in MS lesions [81], with additional evidence indicating that high levels of miR-326 had a strong correlation with increased severity of MS [82]. In two independent studies involving hundreds of SLE patients and healthy controls, decreased expression of miR-146a demonstrated a strong correlation with increased risk for SLE among east Asian and European populations [83], [84]. miRNA expression profiling has also identified type 2 diabetes-related miRNAs including miR-144, miR-146a, miR-150 and miR-182 [85]. In addition, miR-103 and miR-107 were shown to negatively regulate glucose homeostasis and insulin sensitivity in type 2 diabetes by targeting caveolin-1, a critical regulator of insulin receptor [86]. Increased expression of miR-200a, miR-200b and miR-429 and decreased expression of miR-122, miR-451 and miR-27 were associated with diet-induced NAFLD in rats [87]. Furthermore, abnormal expression of miR-29c, miR-34a, miR-155, and miR-200b were found in a mouse model of non-alcoholic steatohepatitis (NASH) [88], in addition to 23 more identified in tissues from NASH patients by miRNA microarrays [89].

Mechanistic studies revealed that miRNAs play critical roles in inflammation primarily by regulating the pathways associated with nuclear factor kappa beta (NF-κB), the central mediator of inflammatory response. The best characterized ones are miR-155 and miR-146, which were implicated in many immune-diseases [73], [74], [81], [85], [88]. In a negative feedback loop in which NF-κB activation up-regulates miR-146 expression, miR-146 subsequently down-regulates the expression of IRAK1 and TRAF6, two up-stream activators of NF-κB [42]. Similarly, increased expression of miR-155 by NF-κB could repress both IKK-β and IKK-ε, and prevent NF-κB from being constitutively activated [90]. This negative feedback mechanism effectively keeps the activity of NF-κB in check. These findings not only provided insights about miRNA-mediated inflammatory responses, but also of potential drug targets for fine-tuning the immune system.

Neurodegenerative diseases

Neurodegenerative diseases (ND) such as Parkinson’s disease (PD) and Alzheimer’s disease (AD) have placed substantial social-economic burdens on countries with aging populations. As the pathogeneses of NDs on molecular levels remain poorly understood, successful treatments are still unavailable. With increasing investments from governments and pharmaceutical companies, biomedical research on neurodegenerative diseases has become proprietary. Notably, recent progresses from studies elucidating miRNA functions in NDs have shed new light on disease pathogenesis and may lead to novel treatment strategies. For example, a systemic miRNA profiling in peripheral blood mononuclear cells from PD patients revealed miR-30b, miR-30c, and miR-26a to be associated with the susceptibility of the disease [91]. Deregulation of miR-133b expression may contribute to the pathogenesis of PD, as the miR-133b-Pitx3 feedback loop is essential for maintaining dopaminergic neurons in the brain [92]. In a Drosophila model for PD, pathogenic leucine-rich repeat kinase 2 (LRRK2) was shown to promote the expression of transcriptional factors E2F1 by down-regulating expression of let-7 and miR-184 [93]. Likewise, an analysis of miRNA and mRNA expression in brain cortex from AD and age-matched control subjects demonstrated strong correlations between the expression levels of miRNAs and predicted mRNA targets [94], implying functional relevance of microRNA-mediated regulations in AD pathogenesis. More specifically, the expression of miR-29a, miR-29b-1 and miR-9 was significantly decreased in AD patients [95], resulting in abnormally high expression of their target BACE1, a protein playing an important role in AD pathogenesis [96]. These findings not only highlight the importance of miRNA research in understanding ND pathogenesis, but also provide a previously unrecognized venue for medical interventions.

miRNAs in disease diagnosis and therapy

While the combination of molecular and computational approaches have revealed the role for miRNAs in common human diseases, concurrent developments of miRNA biomarkers and miRNA drugs have made great strides towards improving public health.

The ultimate goal of biomarker identification is to develop better clinical tests that improve diagnosis or prognosis of diseases. In fact, miRNAs have been considered a top candidate for the next generation of biomarker as they possess a few advantages over other candidates such as proteins and metabolites [97]. First, miRNA biomarkers would more likely lead to early diagnosis due to their upstream positions in regulation cascades. Second, novel miRNA biomarkers would be more readily discovered by genomic tools such as oligonucleotide microarrays and deep sequencing which deliver higher throughput than mass spectrometry, the primary tool for protein and metabolite biomarker identification. Third, low abundant miRNA biomarkers can be amplified and then detected in a clinical setting by real-time quantitative PCR (qPCR), an approach used in FDA-approved clinical tests already; whereas, no equivalent approach is available in detecting low abundant proteins or metabolites. The adoption of the locked-nucleic acid (LNA) technology in miRNA probe design could improve the sensitivity and specificity of miRNA qPCR assays even further [98].

Non-invasive miRNA biomarkers are more sought after due to fewer complications associated with the specimen collection through the more prominent use of bodily fluids such as serum and plasma. In fact, circulating miRNA biomarkers have demonstrated early promises in diagnosis of prostate cancer [99], lung cancer [100], [101], liver cancer [102] and breast cancer [103]. As circulating miRNAs are very stable in the blood [99], [104], they could be well-preserved in archived serum or plasma specimens, a gold mine for miRNA biomarker development.

miRNA drug development is still in its infancy with the exception of SPC3649, a LNA-modified oligonucleotide developed by Santaris Pharma A/S to repress the expression of miR-122, in treating chronic HCV infection. This miRNA drug demonstrated impressive repression efficacy on miR-122 in mice [105] and in African green monkeys [106], as well as anti-viral efficacy in chimpanzees chronically infected by HCV [74]. Compared to a combined administration of pegylated interferon-α and ribavirin, the standard treatment for HCV infection, SPC3649 demonstrated better safety profiles in chimpanzees [74] and desired tolerance in healthy volunteers. Importantly, the SPC3649 treated patients rarely experienced viral-relapse, whereas viral-relapse is common in patients treated with pegylated interferon-α. Interestingly, the expression of interferon-regulated genes decreased in parallel with HCV titers during the SPC3649 treatment. This indicates the effectiveness of SPC3649 on patients infected with viral strains resistant to the interferon-α treatment.

Future directions

In spite of the early success of SPC3649, few miRNA drugs have entered clinical phases due to two major challenges. First, currently available target prediction softwares have high false-positive rates, making it difficult to identify a bona fide miRNA target by in silico prediction alone [107]. To better predict a miRNA drug target before entering costly animal and clinical studies, researchers should take the advantage of combining molecular biology and bioinformatic approaches in target prediction and validation. Recent advancement of molecular biology techniques, such as RISC immune-precipitation and Argonaute-protein crosslinking immune-precipitation, provide valuable tools allowing target enrichment before bioinformatic predications [108], [109]. These techniques should be fully integrated into the studies for target identification. Second, the effective dose of a miRNA drug may induce unsafe off-target effects. A cocktail regimen of miRNAs collaboratively repressing the same target at low doses could be a potential solution. This strategy requires not only extensive bioinformatic efforts in drug designs, but also high-throughput genomic screening to validate the drug effects.

Concluding remarks

Without a doubt, the importance of miRNA is gaining appreciation. However, even with its already demonstrated promise, miRNA diagnosis or therapy may be many years away from entering the clinic as complex challenges remain (Figure 1). It should be noted that any major leap forward in miRNA research over the past decade was the result of multidisciplinary collaborations of researchers with extensive expertise in molecular biology techniques, high-throughput genomics, and bioinformatics. These productive collaborations should be expended even further. With clinicians joining the club, miRNA research will be given a fresh perspective that may lead to steady progress in development of clinical applications.

An external file that holds a picture, illustration, etc.
Object name is gr1.jpg

The road from laboratory to clinic: the promises and challenges of miRNA research The hopscotch course in green is a layout of an ideal path of miRNA research evolved from basic research to clinical practice. Red boxes indicate major challenges at different steps.

Competing interests

The authors declare no conflict of interests.

Acknowledgements

We thank Bryan Maliken for editing assistance. This work is supported by the grants from NIDDK (Grant No. 3R01DK056924-08S1 and 5K24DK002957) and NHLBI (Grant No. 1R21HL112678).

References

1. Bartel D.P. MicroRNAs: genomics, biogenesis, mechanism, and Function. Cell. 2004;116:281–297. [PubMed] [Google Scholar]
2. Lee R.C., Feinbaum R.L., Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75:843–854. [PubMed] [Google Scholar]
3. Reinhart B.J., Slack F.J., Basson M., Pasquinelli A.E., Bettinger J.C., Rougvie A.E. The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature. 2000;403:901–906. [PubMed] [Google Scholar]
4. Kozomara A., Griffiths-Jones S. MiRBase: integrating microRNA annotation and deep-sequencing data. Nucleic Acids Res. 2011;39:D152–D157. [PMC free article] [PubMed] [Google Scholar]
5. Pasquinelli A.E., Reinhart B.J., Slack F., Martindale M.Q., Kuroda M.I., Maller B. Conservation of the sequence and temporal expression of let-7 heterochronic regulatory RNA. Nature. 2000;408:86–89. [PubMed] [Google Scholar]
6. McManus M.T. MicroRNAs and cancer. Semin Cancer Biol. 2003;13:253–258. [PubMed] [Google Scholar]
7. Lagos-Quintana M., Rauhut R., Lendeckel W., Tuschl T. Identification of novel genes coding for small expressed RNAs. Science. 2001;294:853–858. [PubMed] [Google Scholar]
8. Lau N.C., Lim L.P., Weinstein E.G., Bartel D.P. An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science. 2001;294:858–862. [PubMed] [Google Scholar]
9. Lee R.C., Ambros V. An extensive class of small RNAs in Caenorhabditis elegans. Science. 2001;294:862–864. [PubMed] [Google Scholar]
10. Luciano D.J., Mirsky H., Vendetti N.J., Maas S. RNA editing of a miRNA precursor. RNA. 2004;10:1174–1177. [PMC free article] [PubMed] [Google Scholar]
11. Yang W., Chendrimada T.P., Wang Q., Higuchi M., Seeburg P.H., Shiekhattar R. Modulation of microRNA processing and expression through RNA editing by ADAR deaminases. Nat Struct Mol Biol. 2006;13:13–21. [PMC free article] [PubMed] [Google Scholar]
12. Waterston R.H., Lindblad-Toh K., Birney E., Rogers J., Abril J.F., Agarwal P., Mouse Genome Sequencing Consortium Initial sequencing and comparative analysis of the mouse genome. Nature. 2002;420:520–562. [PubMed] [Google Scholar]
13. Gibbs R.A., Rogers J., Katze M.G., Bumgarner R., Weinstock G.M. Evolutionary and biomedical insights from the rhesus macaque genome. Science. 2007;316:222–234. Rhesus Macaque Genome Sequencing and Analysis Consortium. [PubMed] [Google Scholar]
14. Gibbs R.A., Weinstock G.M., Metzker M.L., Muzny D.M., Sodergren E.J., Scherer S. Genome sequence of the Brown Norway rat yields insights into mammalian evolution. Nature. 2004;428:493–521. [PubMed] [Google Scholar]
15. Venter J.C., Adams M.D., Myers E.W., Li P.W., Mural R.J., Sutton G.G. The sequence of the human genome. Science. 2001;291:1304–1351. [PubMed] [Google Scholar]
16. Griffiths-Jones S., Grocock R.J., van Dongen S., Bateman A., Enright A.J. MiRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 2005;34:D140–D144. [PMC free article] [PubMed] [Google Scholar]
17. Bonnet E., Wuyts J., Rouzé P., Van de Peer Y. Evidence that microRNA precursors, unlike other non-coding RNAs, have lower folding free energies than random sequences. Bioinformatics. 2004;20:2911–2917. [PubMed] [Google Scholar]
18. Kim V.N. MicroRNA biogenesis: coordinated cropping and dicing. Nat Rev Mol Cell Biol. 2005;6:376–385. [PubMed] [Google Scholar]
19. Lewis B.P., Burge C.B., Bartel D.P. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120:15–20. [PubMed] [Google Scholar]
20. Chendrimada T.P., Gregory R.I., Kumaraswamy E., Norman J., Cooch N., Nishikura K. TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing. Nature. 2005;436:740–744. [PMC free article] [PubMed] [Google Scholar]
21. Gregory R.I., Chendrimada T.P., Cooch N., Shiekhattar R. Human RISC couples microRNA biogenesis and posttranscriptional gene silencing. Cell. 2005;123:631–640. [PubMed] [Google Scholar]
22. Perron M.P., Provost P. Protein components of the microRNA pathway and human diseases. Methods Mol Biol. 2009;487:369–385. [PMC free article] [PubMed] [Google Scholar]
23. MacRae I.J., Ma E., Zhou M., Robinson C.V., Doudna J.A. In vitro reconstitution of the human RISC-loading complex. Proc Natl Acad Sci U S A. 2008;105:512–517. [PMC free article] [PubMed] [Google Scholar]
24. He L., Hannon G.J. MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet. 2004;5:522–531. [PubMed] [Google Scholar]
25. Lewis B.P., Shih Ih., Jones-Rhoades M.W., Bartel D.P., Burge C.B. Prediction of mammalian microRNA targets. Cell. 2003;115:787–798. [PubMed] [Google Scholar]
26. Krek A., Grun D., Poy M.N., Wolf R., Rosenberg L., Epstein E.J. Combinatorial microRNA target predictions. Nat Genet. 2005;37:495–500. [PubMed] [Google Scholar]
27. John B., Enright A.J., Aravin A., Tuschl T., Sander C., Marks D.S. Human microRNA targets. PLoS Biol. 2004;2:e363. [PMC free article] [PubMed] [Google Scholar]
28. Krüger J., Rehmsmeier M. RNAhybrid: microRNA target prediction easy, fast and flexible. Nucleic Acids Res. 2006;34:W451–W454. [PMC free article] [PubMed] [Google Scholar]
29. Maragkakis M., Reczko M., Simossis V.A., Alexiou P., Papadopoulos G.L., Dalamagas T. DIANA-microT web server: elucidating microRNA functions through target prediction. Nucleic Acids Res. 2009;37:W273–W276. [PMC free article] [PubMed] [Google Scholar]
30. Griffiths-Jones S., Saini H.K., van Dongen S., Enright A.J. MiRBase: tools for microRNA genomics. Nucleic Acids Res. 2008;36:D154–D158. [PMC free article] [PubMed] [Google Scholar]
31. Lim L.P., Lau N.C., Garrett-Engele P., Grimson A., Schelter J.M., Castle J. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature. 2005;433:769–773. [PubMed] [Google Scholar]
32. Sood P., Krek A., Zavolan M., Macino G., Rajewsky N. Cell-type-specific signatures of microRNAs on target mRNA expression. Proc Natl Acad Sci U S A. 2006;103:2746–2751. [PMC free article] [PubMed] [Google Scholar]
33. Huang J.C., Babak T., Corson T.W., Chua G., Khan S., Gallie B.L. Using expression profiling data to identify human microRNA targets. Nat Methods. 2007;4:1045–1049. [PubMed] [Google Scholar]
34. Guo H., Ingolia N.T., Weissman J.S., Bartel D.P. Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature. 2010;466:835–840. [PMC free article] [PubMed] [Google Scholar]
35. Bandiera S., Rüberg S., Girard M., Cagnard N., Hanein S., Chrétien D. Nuclear outsourcing of RNA interference components to human mitochondria. PLoS One. 2011;6:e20746. [PMC free article] [PubMed] [Google Scholar]
36. Beibei C. Bo Zhang, Huaxia Luo, Jiao Yuan, Geir Skogerbø, Runsheng Chen. Distinct microRNA subcellular size and expression patterns in human cancer cells. Int J Cell Biol. 2012;2012:672462. [PMC free article] [PubMed] [Google Scholar]
37. Lagos-Quintana M., Rauhut R., Yalcin A., Meyer J., Lendeckel W., Tuschl T. Identification of tissue-specific microRNAs from mouse. Curr Biol. 2002;12:735–739. [PubMed] [Google Scholar]
38. Lu J., Getz G., Miska E.A., Alvarez-Saavedra E., Lamb J., Peck D. MicroRNA expression profiles classify human cancers. Nature. 2005;435:834–838. [PubMed] [Google Scholar]
39. Ng R., Song G., Roll G.R., Frandsen N.M., Willenbring H. A microRNA-21 surge facilitates rapid cyclin D1 translation and cell cycle progression in mouse liver regeneration. J Clin Invest. 2012;122:1097–1108. [PMC free article] [PubMed] [Google Scholar]
40. Rayner K.J., Esau C.C., Hussain F.N., McDaniel A.L., Marshall S.M., van Gils J.M. Inhibition of miR-33a/b in non-human primates raises plasma HDL and lowers VLDL triglycerides. Nature. 2011;478:404–407. [PMC free article] [PubMed] [Google Scholar]
41. Zhang P., Bill K., Liu J., Young E., Peng T., Bolshakov S. MiR-155 is a liposarcoma oncogene that targets casein kinase-1α and enhances β-catenin signaling. Cancer Res. 2012;72:1751–1762. [PMC free article] [PubMed] [Google Scholar]
42. Taganov K.D., Boldin M.P., Chang K.-J., Baltimore D. NF-κB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci U S A. 2006;103:12481–12486. [PMC free article] [PubMed] [Google Scholar]
43. Png K.J., Halberg N., Yoshida M., Tavazoie S.F. A microRNA regulon that mediates endothelial recruitment and metastasis by cancer cells. Nature. 2012;481:190–194. [PubMed] [Google Scholar]
44. Calin G.A., Dumitru C.D., Shimizu M., Bichi R., Zupo S., Noch E. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A. 2002;99:15524–15529. [PMC free article] [PubMed] [Google Scholar]
45. Calin G.A., Sevignani C., Dumitru C.D., Hyslop T., Noch E., Yendamuri S. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci U S A. 2004;101:2999–3004. [PMC free article] [PubMed] [Google Scholar]
46. Iorio M.V., Ferracin M., Liu C.-G., Veronese A., Spizzo R., Sabbioni S. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005;65:7065–7070. [PubMed] [Google Scholar]
47. Yanaihara N., Caplen N., Bowman E., Seike M., Kumamoto K., Yi M. Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell. 2006;9:189–198. [PubMed] [Google Scholar]
48. Shimizu S., Takehara T., Hikita H., Kodama T., Miyagi T., Hosui A. The let-7 family of microRNAs inhibits Bcl-xL expression and potentiates sorafenib-induced apoptosis in human hepatocellular carcinoma. J Hepatol. 2010;52:698–704. [PubMed] [Google Scholar]
49. Kong W., He L., Coppola M., Guo J., Esposito N.N., Coppola D. MicroRNA-155 regulates cell survival, growth, and chemosensitivity by targeting FOXO3a in breast cancer. J Biol Chem. 2010;285:17869–17879. [PMC free article] [PubMed] [Google Scholar] Retracted
50. Cho W.C.S., Chow A.S.C., Au J.S.K. MiR-145 inhibits cell proliferation of human lung adenocarcinoma by targeting EGFR and NUDT1. RNA Biol. 2011;8:125–131. [PubMed] [Google Scholar]
51. Chan J.A., Krichevsky A.M., Kosik K.S. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res. 2005;65:6029–6033. [PubMed] [Google Scholar]
52. Cimmino A., Calin G.A., Fabbri M., Iorio M.V., Ferracin M., Shimizu M. MiR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci U S A. 2005;102:13944–13949. [PMC free article] [PubMed] [Google Scholar]
53. Johnson S.M., Grosshans H., Shingara J., Byrom M., Jarvis R., Cheng A. RAS is regulated by the let-7 microRNA family. Cell. 2005;120:635–647. [PubMed] [Google Scholar]
54. Valeri N., Gasparini P., Fabbri M., Braconi C., Veronese A., Lovat F. Modulation of mismatch repair and genomic stability by miR-155. Proc Natl Acad Sci U S A. 2010;107:6982–6987. [PMC free article] [PubMed] [Google Scholar]
55. Gao P., Xing A.Y., Zhou G.Y., Zhang T.G., Zhang J.P., Gao C. The molecular mechanism of microRNA-145 to suppress invasion-metastasis cascade in gastric cancer. Oncogene. 2012 http://dx.doi.org/10.1038/onc.2012.61. [PubMed] [Google Scholar]
56. Peng X., Guo W., Liu T., Wang X., Tu Xa, Xiong D. Identification of miRs-143 and -145 that is associated with bone metastasis of prostate cancer and involved in the regulation of EMT. PLoS One. 2011;6:e20341. [PMC free article] [PubMed] [Google Scholar]
57. Volinia S., Galasso M., Sana M.E., Wise T.F., Palatini J., Huebner K. Breast cancer signatures for invasiveness and prognosis defined by deep sequencing of microRNA. Proc Natl Acad Sci U S A. 2012;109:3024–3029. [PMC free article] [PubMed] [Google Scholar]
58. Fang Y.X., Xue J.-L., Shen Q., Chen J., Tian L. MiR-7 inhibits tumor growth and metastasis by targeting the PI3K/AKT pathway in hepatocellular carcinoma. Hepatology. 2012;55:1852–1862. [PubMed] [Google Scholar]
59. Fang J.H., Zhou H.C., Zeng C., Yang J., Liu Y., Huang X. MicroRNA-29b suppresses tumor angiogenesis, invasion, and metastasis by regulating matrix metalloproteinase 2 expression. Hepatology. 2011;54:1729–1740. [PubMed] [Google Scholar]
60. Pfeffer S., Zavolan M., Grässer F.A., Chien M., Russo J.J., Ju J. Identification of virus-encoded microRNAs. Science. 2004;304:734–736. [PubMed] [Google Scholar]
61. Sullivan C.S., Grundhoff A.T., Tevethia S., Pipas J.M., Ganem D. SV40-encoded microRNAs regulate viral gene expression and reduce susceptibility to cytotoxic T cells. Nature. 2005;435:682–686. [PubMed] [Google Scholar]
62. Andersson M.G., Haasnoot P.C.J., Xu N., Berenjian S., Berkhout B., Akusjärvi G. Suppression of RNA interference by adenovirus virus-associated RNA. J Virol. 2005;79:9556–9565. [PMC free article] [PubMed] [Google Scholar]
63. Cai X., Lu S., Zhang Z., Gonzalez C.M., Damania B., Cullen B.R. Kaposi’s sarcoma-associated herpesvirus expresses an array of viral microRNAs in latently infected cells. Proc Natl Acad Sci U S A. 2005;102:5570–5575. [PMC free article] [PubMed] [Google Scholar]
64. Grey F., Antoniewicz A., Allen E., Saugstad J., McShea A., Carrington J.C. Identification and characterization of human cytomegalovirus-encoded microRNAs. J Virol. 2005;79:12095–12099. [PMC free article] [PubMed] [Google Scholar]
65. Pfeffer S., Sewer A., Lagos-Quintana M., Sheridan R., Sander C., Grasser F.A. Identification of microRNAs of the herpesvirus family. Nat Methods. 2005;2:269–276. [PubMed] [Google Scholar]
66. Omoto S., Ito M., Tsutsumi Y., Ichikawa Y., Okuyama H., Brisibe E. HIV-1 nef suppression by virally encoded microRNA. Retrovirology. 2004;1:44. [PMC free article] [PubMed] [Google Scholar]
67. Ouellet D.L., Plante I., Landry P., Barat C., Janelle M.-È., Flamand L. Identification of functional microRNAs released through asymmetrical processing of HIV-1 TAR element. Nucleic Acids Res. 2008;36:2353–2365. [PMC free article] [PubMed] [Google Scholar]
68. Schopman N.C.T., Willemsen M., Liu Y.P., Bradley T., van Kampen A., Baas F. Deep sequencing of virus-infected cells reveals HIV-encoded small RNAs. Nucleic Acids Res. 2012;40:414–427. [PMC free article] [PubMed] [Google Scholar]
69. Lin J., Cullen B.R. Analysis of the interaction of primate retroviruses with the human RNA interference machinery. J Virol. 2007;81:12218–12226. [PMC free article] [PubMed] [Google Scholar]
70. Gatto G., Rossi A., Rossi D., Kroening S., Bonatti S., Mallardo M. Epstein–Barr virus latent membrane protein 1 trans-activates miR-155 transcription through the NF-κB pathway. Nucleic Acids Res. 2008;36:6608–6619. [PMC free article] [PubMed] [Google Scholar]
71. Gottwein E., Mukherjee N., Sachse C., Frenzel C., Majoros W.H., Chi J.-T.A. A viral microRNA functions as an orthologue of cellular miR-155. Nature. 2007;450:1096–1099. [PMC free article] [PubMed] [Google Scholar]
72. Linnstaedt S.D., Gottwein E., Skalsky R.L., Luftig M.A., Cullen B.R. Virally induced cellular microRNA miR-155 plays a key role in B-cell immortalization by Epstein-Barr virus. J Virol. 2010;84:11670–11678. [PMC free article] [PubMed] [Google Scholar]
73. Jopling C.L., Yi M., Lancaster A.M., Lemon S.M., Sarnow P. Modulation of hepatitis C virus RNA abundance by a liver-specific microRNA. Science. 2005;309:1577–1581. [PubMed] [Google Scholar]
74. Lanford R.E., Hildebrandt-Eriksen E.S., Petri A., Persson R., Lindow M., Munk M.E. Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection. Science. 2010;327:198–201. [PMC free article] [PubMed] [Google Scholar]
75. Randall G., Panis M., Cooper J.D., Tellinghuisen T.L., Sukhodolets K.E., Pfeffer S. Cellular cofactors affecting hepatitis C virus infection and replication. Proc Natl Acad Sci U S A. 2007;104:12884–12889. [PMC free article] [PubMed] [Google Scholar]
76. Huang J., Wang F., Argyris E., Chen K., Liang Z., Tian H. Cellular microRNAs contribute to HIV-1 latency in resting primary CD4+ T lymphocytes. Nat Med. 2007;13:1241–1247. [PubMed] [Google Scholar]
77. Li Y., Chan E.Y., Li J., Ni C., Peng X., Rosenzweig E. MicroRNA expression and virulence in pandemic influenza virus-infected mice. J Virol. 2010;84:3023–3032. [PMC free article] [PubMed] [Google Scholar]
78. Li Y., Li J., Belisle S., Baskin C.R., Tumpey T.M., Katze M.G. Differential microRNA expression and virulence of avian, 1918 reassortant, and reconstructed 1918 influenza A viruses. Virology. 2011;421:105–113. [PMC free article] [PubMed] [Google Scholar]
79. Zhang Y., Wei W., Cheng N., Wang K., Li B., Jiang X. Hepatitis C Virus-induced upregulation of miR-155 promotes hepatocarcinogenesis by activating Wnt signaling. Hepatology. 2012;56:1631–1640. [PubMed] [Google Scholar]
80. Keller A., Leidinger P., Lange J., Borries A., Schroers H., Scheffler M. Multiple sclerosis: microRNA expression profiles accurately differentiate patients with relapsing-remitting disease from healthy controls. PLoS One. 2009;4:e7440. [PMC free article] [PubMed] [Google Scholar]
81. Junker A., Krumbholz M., Eisele S., Mohan H., Augstein F., Bittner R. MicroRNA profiling of multiple sclerosis lesions identifies modulators of the regulatory protein CD47. Brain. 2009;132:3342–3352. [PubMed] [Google Scholar]
82. Du C., Liu C., Kang J., Zhao G., Ye Z., Huang S. MicroRNA miR-326 regulates TH-17 differentiation and is associated with the pathogenesis of multiple sclerosis. Nat Immunol. 2009;10:1252–1259. [PubMed] [Google Scholar]
83. Luo X., Yang W., Ye D.-Q., Cui H., Zhang Y., Hirankarn N. A functional variant in microRNA-146a promoter modulates its expression and confers disease risk for systemic lupus erythematosus. PLoS Genet. 2011;7:e1002128. [PMC free article] [PubMed] [Google Scholar]
84. Lofgren S.E., Frostegard J., Truedsson L., Pons-Estel B.A., D’Alfonso S., Witte T. Genetic association of miRNA-146a with systemic lupus erythematosus in Europeans through decreased expression of the gene. Genes Immun. 2012;13:268–274. [PMC free article] [PubMed] [Google Scholar]
85. Karolina D.S., Armugam A., Tavintharan S., Wong M.T.K., Lim S.C., Sum C.F. MicroRNA 144 impairs insulin signaling by inhibiting the expression of insulin receptor substrate 1 in Type 2 Diabetes Mellitus. PLoS One. 2011;6:e22839. [PMC free article] [PubMed] [Google Scholar]
86. Trajkovski M., Hausser J., Soutschek J., Bhat B., Akin A., Zavolan M. MicroRNAs 103 and 107 regulate insulin sensitivity. Nature. 2011;474:649–653. [PubMed] [Google Scholar]
87. Alisi A., Da Sacco L., Bruscalupi G., Piemonte F., Panera N., De Vito R. Mirnome analysis reveals novel molecular determinants in the pathogenesis of diet-induced nonalcoholic fatty liver disease. Lab Invest. 2011;91:283–293. [PubMed] [Google Scholar]
88. Pogribny I.P., Starlard-Davenport A., Tryndyak V.P., Han T., Ross S.A., Rusyn I. Difference in expression of hepatic microRNAs miR-29c, miR-34a, miR-155, and miR-200b is associated with strain-specific susceptibility to dietary nonalcoholic steatohepatitis in mice. Lab Invest. 2010;90:1437–1446. [PMC free article] [PubMed] [Google Scholar]
89. Cheung O., Puri P., Eicken C., Contos M.J., Mirshahi F., Maher J.W. Nonalcoholic steatohepatitis is associated with altered hepatic microRNA expression. Hepatology. 2008;48:1810–1820. [PMC free article] [PubMed] [Google Scholar]
90. Xiao B., Liu Z., Li B.-S., Tang B., Li W., Guo G. Induction of microRNA-155 during Helicobacter pylori infection and its negative regulatory role in the inflammatory response. J Infect Dis. 2009;200:916–925. [PubMed] [Google Scholar]
91. Martins M., Rosa A., Guedes L.C., Fonseca B.V., Gotovac K., Violante S. Convergence of miRNA expression profiling, α-synuclein Interacton and GWAS in Parkinson’s Disease. PLoS One. 2011;6:e25443. [PMC free article] [PubMed] [Google Scholar]
92. Kim J., Inoue K., Ishii J., Vanti W.B., Voronov S.V., Murchison E. A microRNA feedback circuit in midbrain dopamine neurons. Science. 2007;317:1220–1224. [PMC free article] [PubMed] [Google Scholar]
93. Gehrke S., Imai Y., Sokol N., Lu B. Pathogenic LRRK2 negatively regulates microRNA-mediated translational repression. Nature. 2010;466:637–641. [PMC free article] [PubMed] [Google Scholar]
94. Nunez-Iglesias J., Liu C.C., Morgan T.E., Finch C.E., Zhou X.J. Joint genome-wide profiling of miRNA and mRNA expression in Alzheimer’s disease cortex reveals altered miRNA regulation. PLoS One. 2010;5:e8898. [PMC free article] [PubMed] [Google Scholar]
95. Hébert S.S., Horré K., Nicolaï L., Papadopoulou A.S., Mandemakers W., Silahtaroglu A.N. Loss of microRNA cluster miR-29a/b-1 in sporadic Alzheimer’s disease correlates with increased BACE1/β-secretase expression. Proc Natl Acad Sci U S A. 2008;105:6415–6420. [PMC free article] [PubMed] [Google Scholar]
96. Willem M., Garratt A.N., Novak B., Citron M., Kaufmann S., Rittger A. Control of peripheral nerve myelination by the ß-secretase BACE1. Science. 2006;314:664–666. [PubMed] [Google Scholar]
97. Cho W.C.S. MicroRNAs: potential biomarkers for cancer diagnosis, prognosis and targets for therapy. Int J Biochem Cell Biol. 2010;42:1273–1281. [PubMed] [Google Scholar]
98. Kauppinen S., Vester B., Wengel J. Locked nucleic acid: high-affinity targeting of complementary RNA for RNomics. Handb Exp Pharmacol. 2006;173:405–422. [PMC free article] [PubMed] [Google Scholar]
99. Mitchell P.S., Parkin R.K., Kroh E.M., Fritz B.R., Wyman S.K., Pogosova-Agadjanyan E.L. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A. 2008;105:10513–10518. [PMC free article] [PubMed] [Google Scholar]
100. Bianchi F., Nicassio F., Marzi M., Belloni E., Dall’Olio V., Bernard L. A serum circulating miRNA diagnostic test to identify asymptomatic high-risk individuals with early stage lung cancer. EMBO Mol Med. 2011;3:495–503. [PMC free article] [PubMed] [Google Scholar]
101. Hennessey P.T., Sanford T., Choudhary A., Mydlarz W.W., Brown D., Adai A.T. Serum microRNA biomarkers for detection of non-small cell lung cancer. PLoS One. 2012;7:e32307. [PMC free article] [PubMed] [Google Scholar]
102. Qi P., Cheng S.-Q., Wang H., Li N. Chen Y-f, Gao C-f. Serum microRNAs as biomarkers for hepatocellular carcinoma in Chinese patients with chronic hepatitis B virus infection. PLoS One. 2011;6:e28486. [PMC free article] [PubMed] [Google Scholar]
103. Roth C., Rack B., Muller V., Janni W., Pantel K., Schwarzenbach H. Circulating microRNAs as blood-based markers for patients with primary and metastatic breast cancer. Breast Cancer Res. 2010;12:R90. [PMC free article] [PubMed] [Google Scholar]
104. Arroyo J.D., Chevillet J.R., Kroh E.M., Ruf I.K., Pritchard C.C., Gibson D.F. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci U S A. 2011;108:5003–5008. [PMC free article] [PubMed] [Google Scholar]
105. Elmén J., Lindow M., Silahtaroglu A., Bak M., Christensen M., Lind-Thomsen A. Antagonism of microRNA-122 in mice by systemically administered LNA-antimiR leads to up-regulation of a large set of predicted target mRNAs in the liver. Nucleic Acids Res. 2008;36:1153–1162. [PMC free article] [PubMed] [Google Scholar]
106. Elmen J., Lindow M., Schutz S., Lawrence M., Petri A., Obad S. LNA-mediated microRNA silencing in non-human primates. Nature. 2008;452:896–899. [PubMed] [Google Scholar]
107. Yue D., Liu H., Huang Y. Survey of computational algorithms for microRNA target prediction. Curr Genomics. 2009;10:478–492. [PMC free article] [PubMed] [Google Scholar]
108. Karginov F.V., Conaco C., Xuan Z., Schmidt B.H., Parker J.S., Mandel G. A biochemical approach to identifying microRNA targets. Proc Natl Acad Sci U S A. 2007;104:19291–19296. [PMC free article] [PubMed] [Google Scholar]
109. Chi S.W., Zang J.B., Mele A., Darnell R.B. Argonaute HITS-CLIP decodes microRNA-mRNA interaction maps. Nature. 2009;460:479–486. [PMC free article] [PubMed] [Google Scholar]

Articles from Genomics, Proteomics & Bioinformatics are provided here courtesy of Oxford University Press

-