Vous êtes sur la page 1sur 5

Gene 451 (2010) 15

Contents lists available at ScienceDirect

Gene
j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / g e n e

Review

Experimental identication of microRNA targets


Ulf Andersson rom 1, Anders H. Lund
Biotech Research and Innovation Centre and Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark

a r t i c l e

i n f o

a b s t r a c t
microRNAs are small RNAs that regulate protein synthesis post-transcriptionally. Animal microRNAs recognize their targets by incomplete base pairing to sequence motifs most often present in the 3 untranslated region of their target mRNAs. This partial complementarity vastly expands the repertoire of potential targets and constitutes a problem for computational target prediction. Although computational analyses have shed light on important aspects of microRNA target recognition, several questions remain regarding how microRNAs can recognize and regulate their targets. Forward experimental approaches allow for an unbiased study of microRNA target recognition and may unveil novel, rare or uncommon target binding patterns. In this review we focus on animal microRNAs and the experimental approaches that have been described for identication of their targets. 2009 Elsevier B.V. All rights reserved.

Article history: Received 8 October 2009 Received in revised form 10 November 2009 Accepted 16 November 2009 Available online 24 November 2009 Received by A. J. Van Wijren Keywords: microRNA Target identication

1. Introduction microRNAs (miRNAs) are uncapped, unpolyadenylated small RNAs that are processed from primary transcripts in sequential steps by the RNase III endonucleases Drosha in the nucleus (Lee et al., 2003) and Dicer in the cytoplasm (Hutvagner, 2005). Mature miRNA are incorporated into the RNA-induced silencing complex (RISC; Meister et al., 2004b) where they are bound by members of the Argonaute (Ago) family of proteins and constitute the target recognition module of RISC (Carthew and Sontheimer, 2009). Extensive research has revealed the existence of more than 700 different human miRNAs (Grifths-Jones et al., 2008) and numerous reports have demonstrated the importance of miRNA-mediated regulation in key processes, such as proliferation, apoptosis, differentiation and development, cellular identity and pathogenhost interactions (He et al., 2007; Parker and Sheth, 2007; Pillai et al., 2007; Carthew and Sontheimer, 2009). Despite of this, the mechanisms by which miRNAs act are still not resolved. The rst step toward unraveling the function of a particular miRNA is the identication of its direct targets. This step has proven to be quite challenging in animals primarily due to the incomplete complementarity between miRNA and target mRNAs. Some key principles have emerged on the pattern of miRNA target recognition and these have been applied to computationally predict targets of miRNA regulation (Bartel, 2009). Examples of commonly

Abbreviations: UTR, untranslated region; miRNA, microRNA; RISC, RNA-induced silencing complex; SILAC, stable isotope labeling by amino acids in cell culture; HITS-CLIP, high-throughput sequencing of RNAs isolated by cross-linking immunoprecipitation. Corresponding author. E-mail address: anders.lund@bric.ku.dk (A.H. Lund). 1 Present address: The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA. 0378-1119/$ see front matter 2009 Elsevier B.V. All rights reserved. doi:10.1016/j.gene.2009.11.008

used algorithms are miRanda (John et al., 2004), TargetScan (Lewis et al., 2003, 2005) and PicTar (Krek et al., 2005). The most general feature of miRNA regulation described is the recognition of sequence motifs complementary to the seed region (nucleotides 27 of the miRNA) in the 3 UTR of target mRNAs (Lewis et al., 2003), which together with criteria such as target sequence conservation make up the basis for most target prediction algorithms. It is currently unknown which proportion of miRNA interactions follow these rules and functional recognition motifs outside of the 3 UTRs, not following the seed rule and target sequences that are not conserved between species, have been reported (Ha et al., 1996; Reinhart and Bartel, 2002; Vella et al., 2004b; Jopling et al., 2005; Krek et al., 2005; Didiano and Hobert, 2006; Easow et al., 2007; Orom et al., 2008; Tay et al., 2008; Tsai et al., 2009). Computational approaches to miRNA target identication are strong tools to narrow down the list of putative targets of miRNA regulation and have contributed signicantly to the development of the miRNA eld. However, a limitation of target predictions is that they rely on few established principles and as such cannot help in revealing novel aspects of miRNA target recognition. While several reports document the validity of predicted targets for miRNA regulation, many predicted targets do not recapitulate regulation in validation experiments (Nakamoto et al., 2005; Vinther et al., 2006; Frankel et al., 2008; Baek et al., 2008; Didiano and Hobert, 2008; Selbach et al., 2008; Jiang et al., 2009). A thorough study of miRNAs predicted to target CyclinD1 has addressed this using luciferase reporter assays (Jiang et al., 2009). Out of 45 miRNAs predicted to target the CyclinD1 3 UTR only 7 could be conrmed by the authors (16%). While false positive predictions can be eliminated by experimental validation studies, the number of false negative predictions remains unknown. An unbiased approach to study miRNA interactions with their targets would provide much insight

U.A. rom, A.H. Lund / Gene 451 (2010) 15

into additional recognition patterns and help as well to exclude false negative predictions. In this review, we describe the reported experimental approaches to identify the mRNA targets associated with specic miRNAs in animals (for overview, see Fig. 1). 2. Experimental target identication 2.1. Transcriptome analyses The realization that animal miRNAs down-regulate the level of a number of their target mRNAs (Bagga et al., 2005; Lim et al., 2005) paved the way for a series of overexpression and miRNA inhibition studies where miRNA targets were sought identied on a transcriptome-wide scale (Krutzfeldt et al., 2005; Christoffersen et al., 2007; Frankel et al., 2008; Grimson et al., 2007; Elmen et al., 2008a). Initial studies transiently transfected the tissue specic miRNAs miR-1 (muscle specic) and miR-124a (brain specic) into HeLa cells where they are normally not expressed and used microarray analyses to identify the cohort of mRNAs down-regulated as a consequence of miRNA overexpression (Lim et al., 2005). Subsequent analysis showed that target mRNA down-regulation is highly signicantly associated with the presence of an miRNA seed complementary site in the mRNA 3 UTR sequence. In addition, correlations between the mRNA targets and the miRNAs are shown: identied targets are primarily expressed at low levels in the tissues with high expression of the miRNAs (Farh et al., 2005; Lim et al., 2005). Furthermore, introducing the tissue specic miRNAs into HeLa cells shifted the mRNA expression prole toward that of the tissue normally expressing the miRNA, suggesting a very important role for miRNAs in tissue development and maintenance (Lim et al., 2005). The option to identify a large set of miRNA targets using microarrays has prompted other groups to take similar approaches to unravel miRNA functions both in cell culture and in vivo. A modied approach, in part trying to avoid offtarget effects resulting from miRNA overexpression, is to inhibit the miRNA of interest with oligonucleotides complementary to the

miRNA (Hutvagner et al., 2004; Meister et al., 2004a; Orom et al., 2006) and analyze mRNA levels on microarrays. When inhibiting the miRNA a subset of its targets will increase at both the protein and mRNA levels and potential targets can thus be readily identied (Krutzfeldt et al., 2005; Frankel et al., 2008ff; Elmen et al., 2008b; Christoffersen et al., 2009). Two reports apply both overexpression and inhibition of miRNAs (Nicolas et al., 2008; Ziegelbauer et al., 2009). By analyzing the overlap between these two series of experiments the list of putative direct target is signicantly reduced. When miR-140 was either overexpressed or inhibited (Nicolas et al., 2008) a list of 1236 and 466 genes were reported as differentially expressed, while the overlap between the two experiments was only 49 transcripts. Twenty-one of these 49 mRNAs contain miR-140 seed complementary sites, yet none of them are predicted by commonly used miRNA target prediction algorithms, suggesting a signicant number of false negative predictions by these algorithms. While these approaches can identify a subset of miRNA targets, they are limited to the mRNAs that are degraded to a certain extent by their targeting miRNAs, and the applications of such approaches have been highly dependent on computational analyses based on sequence complementarity. Such an approach yields many candidate target mRNAs that are differentially expressed upon exogenous introduction of miRNAs and most likely many false positive candidates are included due to downstream effects of the affected true miRNA mRNA targets. An approach to limit the number of false positives is to rely on seed site complementarity in the detected candidates. It is evident from these experiments that destabilization of target mRNAs is an important mechanism for miRNA function, on top of the strict translational repression without effects on mRNA levels. 2.2. Biochemical approaches Several known miRNA targets have been identied using bioinformatic analyses for seed complementarity and subsequent experimental and functional validation of the interaction. A more challenging task is to identify those targets regulated primarily at the level of translation, or recognized through non-seed base pairing interactions. Toward this, several groups have reported progress using different experimental approaches. Three reports address experimental miRNA target identication by immunoprecipitation of Ago proteins, either tagged or endogenous, to analyze the associated mRNAs as candidate miRNA targets. Karginov et al. used an epitope-tagged Ago2 in HEK293 to isolate targets of mir-124a, an miRNA not endogenously expressed in HEK293 cells (Karginov et al., 2007). Initial validation of the approach showed signicant enrichment of three previously characterized targets of miR-124a, Ctdsp1, Plod3 and Vamp3, whereas a panel of housekeeping mRNAs was not enriched after immunoprecipitation of the myc-tagged Ago2. To identify a comprehensive set of miR-124a targets the myc-Ago2 immunoprecipitates were hybridized to microarrays along with determination of total mRNA levels. Both mRNA targets that are down-regulated in total mRNA and targets that are unaffected at the mRNA level by the miRNA were identied in the immunoprecipitates. Four of 4 down-regulated mRNA targets and 21 of 30 tested mRNAs that were not affected at total mRNA level were validated in luciferase reporter 3 UTR assays, but a further characterization of the translationally regulated targets was not pursued. The paper shows that miRNA targets can be isolated and identied using Ago immunoprecipitation, identifying primarily those targets that are translationally repressed. Similar ndings were demonstrated for miR-1 in a Drosophila system (Easow et al., 2007). Using immunoprecipitation of HA-tagged Ago1 proteins in S2 cells and subsequent microarray analysis, enrichments for mRNAs containing miR-1 miRNA seed complementary sites in their 3 UTRs were demonstrated to correlate with the expression level of the specic

Fig. 1. Overview of approaches for experimentally identifying microRNA targets. microRNA regulation of translation is a multi-facetted process that allows several entrances for experimentally identifying the targets regulated by a specic microRNA. Reports address this issue through: (1) Analysis of mRNAs degraded as a consequence of overexpressing the microRNA and subsequent analysis of sequence motifs, (2) immunoprecipitation of tagged or endogenous RISC complex and analysis of associated mRNAs, (3) Afnity purication of tagged microRNAs and microarray analysis of associated mRNAs, (4) by using the observation that some microRNA targets move in the polysomal distribution upon microRNA targeting and analyzing differences in polysomal associated mRNAs with and without the microRNA, (5) analyzing protein production following labeling of proteins and mass spectrometry.

U.A. rom, A.H. Lund / Gene 451 (2010) 15

miRNAs. The study shows as well the applicability of Ago immunoprecipitation for miRNA target identication, but lacks a thorough analysis of the identied targets. Rather the report focuses on the presence of miR-1 seeds in a subset of the identied potential targets of miR-1 regulation. Beitzinger et al. (2007) isolated endogenous Ago proteins from HEK293 cells using highly specic monoclonal antibodies against either human Ago1 or human Ago2 (Beitzinger et al., 2007). By purifying RNAs associated with either of the Ago proteins, cDNA synthesis and cloning, the associated mRNAs were identied. Analysis of the putative miRNA targets shows little overlap between Ago1- and Ago2-associated miRNA targets in human HEK293 cells, suggesting that specic pools of miRNAs or miRNA targets are associated to the different Ago proteins. About half of the suggested targets were predicted by at least one of the three applied target prediction methods: MiRanda (John et al., 2004), TargetScan (Lewis et al., 2003, 2005) or Pictar (Krek et al., 2005). For validation, 6 mRNAs predicted to be targets of miRNA regulation were selected. Cloning of their 3 UTRs into a luciferase reporter vector and reporter assays with both miRNA overexpression or miRNA inhibition conrmed that these targets are regulated by the predicted miRNA through their 3 UTRs. While all three studies report the identication of miRNA targets using experimental approaches, none of them address miRNA target recognition directly but tend to rely on miRNA seed site interaction for validation. The three papers show the potential of Ago immunoprecipitation as a means of identifying miRNA targets but at the same time they demonstrate the inherited difculties in experimental miRNA target identication. While several thousands of mRNAs are hypothesized to be regulated by miRNAs, only a few are identied using these approaches. Tagging of the miRNA is another approach that has been employed to identify targets of miRNA regulation. By transfecting cells with miRNAs labeled with biotin and subsequently isolating the associated mRNAs, this method has been described for the well-characterized bantam/hid interaction in Drosophila both in reporter assays in HEK293 cells and for endogenous hid in S2 cells, where the hid 3 UTR could be afnity puried using a biotin-tagged bantam miRNA (Orom and Lund, 2007). The method has been used to validate individual miRNA:target interactions (Kedde et al., 2007; Christoffersen et al., 2009) and to identify targets and suggest a novel function of the miRNA miR-10a (Orom et al., 2008). Surprisingly, it was found that miRNA-10a can target mRNAs encoding ribosomal proteins through their 5 UTRs via non-seed interactions to enhance their translation, as well as modulate mRNA targets through their 3 UTRs and repress their translation (Orom et al., 2008). Using this method, it was shown by cross-linking followed by primer extension mapping of the miRNA binding site that the non-canonical interaction is direct, which is also validated by mutating the miRNA target sequence and the corresponding bases in the miRNA to recover the enhancing effect observed of the miRNA. An in vitro procedure using digoxigenin-labeled miRNA precursors has also been employed (Hsu et al., 2009). By incubation with antiDIG antiserum known miRNA targets from C. elegans and zebrash were conrmed using qPCR. Additionally the approach identied hand2 as a miR-1 target. Controversy exists about miRNA target association to polysomes. mRNAs targeted by miRNAs are both reported associated to polysomes while bound by miRNAs and reported to shuttle in the polysomal spectrum as a consequence of miRNA regulation (Olsen and Ambros 1999; Nelson et al., 2004; Nakamoto et al., 2005; Pillai et al., 2005; Petersen et al., 2006; Thermann and Hentze, 2007). Nakamoto et al. have used the assumption that the position of a transcript in a polysome prole reects, in part, the degree of its translation. Hence, shifts into heavier polysome fractions would reect increased translation (Nakamoto et al., 2005). Using knockdown of endogenous miR-30a-3p and isolating polysomal and sub-

polysomal fractions and comparing associated mRNAs on microarrays, 8 mRNAs translationally induced upon miR-30a-3p knockdown were identied and validated as being targets of miR-30a-3p regulation. Despite that all 8 mRNAs contain seed sites (including G:U wobble pairs), none of them were predicted to be targets of miR-30a3p by the applied algorithms with a score above threshold. This study clearly demonstrates the applicability of forward approaches to identify miRNA targets. Even though only a few target candidates are identied, none of them were previously predicted to be targets of miR-30a-3p. A recent report using purication of cross-linked RNA-binding proteins has shed more light on miRNA target recognition (Chi et al., 2009). This approach, termed HITS-CLIP, uses ultraviolet light to cross-link Ago proteins to associated RNA and miRNA. Ago protein complexes were immunoprecipitated and puried from mouse brains and the associated RNA identied by sequencing. Clusters of Ago binding sites were then identied, which provided not only the bound transcript but also the position of Ago binding. The study identies 1463 Ago clusters mapping to 829 transcripts. The identity of the miRNA bound to each target is not known with this approach. The authors use bioinformatics prediction to account for their presumption that the 20 most expressed miRNAs account for the majority of bound targets, however 27% of identied targets do not contain sequences corresponding to the 20 most expressed miRNAs. miRNAs are shown to bind mostly to 3 UTRs but also to a large degree to the open reading frames of the identied targets, although it is unclear if these binding sites are functional. The brain specic miRNA miR-124 was used to compare to bioinformatics predictions for miR-124 targets. Interestingly, there is a substantial overlap between targets identied for miR-124 using HITS-CLIP and computationally predicted transcripts, although the experimental approach identies fewer binding sites for Agos in each transcript. This study provides insight on miRNA target recognition and can potentially assist in unraveling as yet uncharacterized patterns of miRNA target recognition, as the approach not only can help identify the targets of miRNA regulation but also dene the region within which the interaction takes place. The option of studying single miRNAs with this approach would give even more insightful knowledge on the target recognition properties of a single miRNA without having to guess some of the interactions or make assumptions of which miRNAs are binding the identied target mRNA. Currently, further development of this method is ongoing in several laboratories. 2.3. Proteome analyses Several proteomic approaches for studying miRNA target regulation using stable isotope labeling by amino acids in cell culture (SILAC) have been reported (Vinther et al., 2006; Baek et al., 2008; Selbach et al., 2008). This experimental approach is appealing as it may identify targets regulated both by transcript destabilization and translational repression. With SILAC, proteins are metabolically labeled by growing cells in medium containing heavy isotopes of essential amino acids typically lysine and arginine. Using mass spectrometry, differences in protein synthesis can be determined by the ratio of peptide peak intensities from the light and heavy isotopes. The rst study to apply SILAC for miRNA target identication found 12 targets for the miRNA miR-1 in HeLa cells (Vinther et al., 2006). Eight of the 12 identied targets contain seed complementary sites in their 3 UTRs. A comparison with mRNA microarray analysis studies of miR-1 targets in HeLa cells (Lim et al., 2005) showed that four of these targets overlap between the two studies using different approaches to address the same question. Luciferase reporter validation of 3 UTRs of the identied target genes supported 6 of the putative target mRNAs identied, underlining the applicability of the method for miRNA target identication. Following this report, two large-scale proteomics studies to identify miRNA targets have been published (Baek et al.,

U.A. rom, A.H. Lund / Gene 451 (2010) 15

2008; Selbach et al., 2008). Baek et al. studied the miRNAs miR-1, miR124 and miR-181 in HeLa cells and the effect of removing miR-223 in mouse neutrophils. Selbach et al. used a slightly modied SILAC procedure where cells were pulse-labeled to incorporate the isotopes primarily into newly synthesized proteins, and studied the miRNAs miR-1, miR-30, miR-155, miR-16 and let-7b and knock-down of let-7b in HeLa cells. While one large-scale study reports primarily effects at the level of mRNA stability (Baek et al., 2008), another observes more instances of specic translational inhibition (Selbach et al., 2008). Common to the two reports is that they show effects of single miRNAs on hundreds of proteins, albeit with a bias toward the detection of proteins expressed at a higher level. Most of these effects are modest, making it hard to distinguish primary miRNA effects from secondary effects. Analyses for predicted binding sites in the 3 UTRs show enrichment for the presence of seed sites. The small effects observed lead the authors to suggest that an important role of miRNAs might be the ne-tuning of the expression of many proteins. In addition, several putative targets show up-regulation of protein synthesis, suggesting a general enhancing effect of miRNAs (Selbach et al., 2008), either indirect or direct, on a large number of proteins. An example of a clinically applicable small-scale proteomics approach using reverse-phase protein miRNA analysis has been described (Iliopoulos et al., 2008). Comparison of miRNA expression and reverse-phase protein arrays probed with 214 antibodies in combination with miRNA target prediction identied a number of putative targets of miRNA regulation involved in the pathogenesis of osteoarthritis. The study identied and validated the regulation by miR-22 of BMP7 and PPARa. While the approach relies completely on target prediction algorithms, it is advantageous for analysis of clinical samples where the amount of sample is limited. 3. Discussion When considering the several approaches reported to successfully identify mRNA targets of miRNA regulation only few experimentally identied and functionally validated miRNA targets exist. This likely reects the challenge of miRNA target identication and subsequent useful functional validation. miRNA target validation focusing on computationally predicted targets has been discussed recently (Kuhn et al., 2008; Bartel, 2009). For experimentally identied targets, functional validation is more relevant than computational analyses. Approaches such as calculation of G values are mostly useful to narrow down the number of putative candidate target mRNAs from bioinformatics analyses and may also exclude true targets. Effects on endogenous target protein levels serve as good indicators for valid miRNA target interactions, although indirect effects cannot be excluded from these experiments. A more direct validation, although not in its natural context, can be obtained by cloning a sequence of the mRNA of interest into a luciferase reporter and do co-transfection reporter assays. By mutating the identied target site and subsequently introducing complementary mutations into the miRNA sequence, abrogation and restoration of the translational effect on the reporter should be observed for a true miRNA target. This approach suffers from the limitation that both target and miRNA are present at articially high concentrations, which may affect the effect observed (Doench and Sharp, 2004). Furthermore, direct evidence that an mRNA is endogenously bound by an miRNA can be obtained by using either formaldehyde cross-linking of the miRNA to its targets (Vasudevan et al., 2007) or 4-thiouridine-modied miRNAs (Orom et al., 2008), that allows for subsequent mapping of the exact site of binding using primer extension. The data obtained from experimental approaches to identify miRNA targets should, in addition to identifying targets involved in the processes studied, be used to characterize miRNA binding patterns

further. Most of the approaches described in this review resort to using the proposed seed pattern of miRNA recognition of their targets as a validation criterion for the success of their approach, rather than asking which patterns of recognition can be deduced from their data. Flanking sequences outside of the miRNA recognition site have been suggested to have important regulatory functions for a number of miRNAs (Vella et al., 2004a; Didiano and Hobert, 2006; Grimson et al., 2007; Kertesz et al., 2007; Didiano and Hobert, 2008), but very little has been done so far toward identifying additional mRNA determinants for miRNA binding and function. A major problem with an unbiased forward approach in target site analysis is the rather limited number of experimentally identied and validated targets each approach has revealed. With the recent, largescale proteomic approaches, together with genome-wide mapping of miRNA binding regions coming from techniques such as HITS-CLIPS, this may no longer be a limitation. 4. Conclusion Identifying targets of miRNA regulation remains a fundamental challenge and the lack of knowledge concerning the different mechanisms by which miRNAs work constitutes a major problem for experimental target identication. Hence, a combination of target identication methods may turn out to be necessary to reveal the full spectrum of miRNA target regulation. While the approaches applying Ago tagging and immunoprecipitation will likely miss degraded mRNAs, these are readily picked up by transfection and microarray approaches, which in turn cannot be used to identify targets that are exclusively regulated at the level of translation. The most comprehensive approach described so far for miRNA target identication is the proteomics approach reported by three different groups (Vinther et al., 2006; Baek et al., 2008; Selbach et al., 2008), and such an approach should be able to pick up all kinds of repression by the miRNA, as the output is protein levels. While it remains problematic to distinguish primary and secondary effects without relying on extensive experimental validation or on computational predictions, global proteomics approaches could reveal new aspects of miRNA target site recognition and function. While repression is by far the most commonly reported effect of miRNA targeting of an mRNA, enhancement of translation by miRNAs has been observed by a handful of groups so far (Vasudevan et al., 2007; Henke et al., 2008; Orom et al., 2008; Selbach et al., 2008; Iwasaki and Tomari, 2009; Tsai et al., 2009), two of which are based on experimental target identication. This could be a consequence of different miRNA recognition motifs, of mRNA sequence context, or as recently suggested due to cell cycle-dependent differences in miRNA functions (Vasudevan et al., 2007). In summary, experimental identication of miRNA targets should to a higher extent be used to expand the current knowledge of miRNA target recognition and broadening of the spectrum of miRNA targets. Acknowledgments Work in the authors' laboratory is supported by EC FP7 funding (ONCOMIRS, Grant Agreement Number 201102. This publication reects only the authors' views. The commission is not liable for any use that may be made of the information herein), the Novo Nordisk Foundation, the Danish National Research Foundation, the Danish Medical Research Council, the Danish Cancer Society and the Danish National Advanced Technology Foundation. UA is supported by a personal grant from the Danish Medical Research Council. References
Baek, D., Villen, J., Shin, C., Camargo, F.D., Gygi, S.P., Bartel, D.P., 2008. The impact of microRNAs on protein output. Nature 455, 6471.

U.A. rom, A.H. Lund / Gene 451 (2010) 15 Bagga, S., et al., 2005. Regulation by let-7 and lin-4 miRNAs results in target mRNA degradation. Cell 122, 553563. Bartel, D.P., 2009. MicroRNAs: target recognition and regulatory functions. Cell 136, 215233. Beitzinger, M., Peters, L., Zhu, J.Y., Kremmer, E., Meister, G., 2007. Identication of human microRNA targets from isolated argonaute protein complexes. RNA Biol. 4, 7684. Carthew, R.W., Sontheimer, E.J., 2009. Origins and mechanisms of miRNAs and siRNAs. Cell 136, 642655. Chi, S.W., Zang, J.B., Mele, A., Darnell, R.B., 2009. Argonaute HITS-CLIP decodes microRNAmRNA interaction maps. Nature 460, 479486. Christoffersen, N.R., et al., 2009. p53-independent upregulation of miR-34a during oncogene-induced senescence represses MYC. Cell Death and Differentiation. doi:10.1038/cdd.2009.109. Christoffersen, N.R., Silahtaroglu, A., Orom, U.A., Kauppinen, S., Lund, A.H., 2007. miR-200b mediates post-transcriptional repression of ZFHX1B. Rna 13, 11721178. Didiano, D., Hobert, O., 2006. Perfect seed pairing is not a generally reliable predictor for miRNA-target interactions. Nat. Struct. Mol. Biol. 13, 849851. Didiano, D., Hobert, O., 2008. Molecular architecture of a miRNA-regulated 3 UTR. Rna 14, 12971317. Doench, J.G., Sharp, P.A., 2004. Specicity of microRNA target selection in translational repression. Genes Dev. 18, 504511. Easow, G., Teleman, A.A., Cohen, S.M., 2007. Isolation of microRNA targets by miRNP immunopurication. Rna 13, 11981204. Elmen, J., et al., 2008a. LNA-mediated microRNA silencing in non-human primates. Nature 452, 896899. Elmen, J., et al., 2008b. 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. 36, 11531162. Farh, K.K., Grimson, A., Jan, C., Lewis, B.P., Johnston, W.K., Lim, L.P., Burge, C.B., Bartel, D.P., 2005. The widespread impact of mammalian MicroRNAs on mRNA repression and evolution. Science 310, 18171821. Frankel, L.B., Christoffersen, N.R., Jacobsen, A., Lindow, M., Krogh, A., Lund, A.H., 2008. Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J. Biol. Chem. 283, 10261033. Grifths-Jones, S., Saini, H.K., Dongen, S.V., Enright, A.J., 2008. miRBase: tools for microRNA genomics. Nucleic Acids Res. 36, D154D158. Grimson, A., Farh, K.K., Johnston, W.K., Garrett-Engele, P., Lim, L.P., Bartel, D.P., 2007. MicroRNA targeting specicity in mammals: determinants beyond seed pairing. Mol. Cell 27, 91105. Ha, I., Wightman, B., Ruvkun, G., 1996. A bulged lin-4/lin-14 RNA duplex is sufcient for Caenorhabditis elegans lin-14 temporal gradient formation. Genes Dev. 10, 30413050. He, L., He, X., Lowe, S.W., Hannon, G.J., 2007. microRNAs join the p53 networkanother piece in the tumour-suppression puzzle. Nat. Rev. Cancer 7, 819822. Henke, J.I., et al., 2008. microRNA-122 stimulates translation of hepatitis C virus RNA. EMBO J. 27, 33003310. Hsu, R.J., Yang, H.J., Tsai, H.J., 2009. Labeled microRNA pull-down assay system: an experimental approach for high-throughput identication of microRNA-target mRNAs. Nucleic Acids Res. 37, e77. Hutvagner, G., 2005. Small RNA asymmetry in RNAi: function in RISC assembly and gene regulation. FEBS Lett. 579, 58505857. Hutvagner, G., Simard, M.J., Mello, C.C., Zamore, P.D., 2004. Sequence-specic inhibition of small RNA function. PLoS Biol. 2, E98. Iliopoulos, D., Malizos, K.N., Oikonomou, P., Tsezou, A., 2008. Integrative microRNA and proteomic approaches identify novel osteoarthritis genes and their collaborative metabolic and inammatory networks. PLoS One 3, e3740. Iwasaki, S., Tomari, Y., 2009. Argonaute-mediated translational repression (and activation). Fly (Austin) 3, 204206. Jiang, Q., Feng, M., Mo, Y.Y., 2009. Systematic validation of predicted microRNAs for cyclin D1. BMC Cancer 9, 194. John, B., Enright, A.J., Aravin, A., Tuschl, T., Sander, C., Marks, D.S., 2004. Human MicroRNA targets. PLoS Biol. 2, e363. Jopling, C.L., Yi, M., Lancaster, A.M., Lemon, S.M., Sarnow, P., 2005. Modulation of hepatitis C virus RNA abundance by a liver-specic MicroRNA. Science 309, 15771581. Karginov, F.V., et al., 2007. A biochemical approach to identifying microRNA targets. Proc. Natl. Acad. Sci. U S A 104, 1929119296. Kedde, M., et al., 2007. RNA-binding protein Dnd1 inhibits microRNA access to target mRNA. Cell 131, 12731286.

Kertesz, M., Iovino, N., Unnerstall, U., Gaul, U., Segal, E., 2007. The role of site accessibility in microRNA target recognition. Nat. Genet. 39, 12781284. Krek, A., et al., 2005. Combinatorial microRNA target predictions. Nat. Genet. 37, 495500. Krutzfeldt, J., et al., 2005. Silencing of microRNAs in vivo with antagomirs. Nature 438, 685689. Kuhn, D.E., Martin, M.M., Feldman, D.S., Terry, A.V., Jr., Nuovo, G.J., Elton, T.S., 2008. Experimental validation of miRNA targets. Methods 44, 4754. Lee, Y., et al., 2003. The nuclear RNase III Drosha initiates microRNA processing. Nature 425, 415419. Lewis, B.P., Burge, C.B., Bartel, D.P., 2005. Conserved seed pairing, often anked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120, 1520. Lewis, B.P., Shih, I.H., Jones-Rhoades, M.W., Bartel, D.P., Burge, C.B., 2003. Prediction of mammalian microRNA targets. Cell 115, 787798. Lim, L.P., et al., 2005. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature 433, 769773. Meister, G., Landthaler, M., Dorsett, Y., Tuschl, T., 2004a. Sequence-specic inhibition of microRNA- and siRNA-induced RNA silencing. Rna 10, 544550. Meister, G., Landthaler, M., Patkaniowska, A., Dorsett, Y., Teng, G., Tuschl, T., 2004b. Human Argonaute2 mediates RNA cleavage targeted by miRNAs and siRNAs. Mol. Cell 15, 185197. Nakamoto, M., Jin, P., O'Donnell, W.T., Warren, S.T., 2005. Physiological identication of human transcripts translationally regulated by a specic microRNA. Hum. Mol. Genet. 14, 38133821. Nelson, P.T., Hatzigeorgiou, A.G., Mourelatos, Z., 2004. miRNP:mRNA association in polyribosomes in a human neuronal cell line. Rna 10, 387394. Nicolas, F.E., et al., 2008. Experimental identication of microRNA-140 targets by silencing and overexpressing miR-140. Rna 14, 25132520. Olsen, P.H., Ambros, V., 1999. The lin-4 regulatory RNA controls developmental timing in Caenorhabditis elegans by blocking LIN-14 protein synthesis after the initiation of translation. Dev Biol 216, 671680. Orom, U.A., Kauppinen, S., Lund, A.H., 2006. LNA-modied oligonucleotides mediate specic inhibition of microRNA function. Gene 372, 137141. Orom, U.A., Lund, A.H., 2007. Isolation of microRNA targets using biotinylated synthetic microRNAs. Methods 43, 162165. Orom, U.A., Nielsen, F.C., Lund, A.H., 2008. MicroRNA-10a binds the 5UTR of ribosomal protein mRNAs and enhances their translation. Mol. Cell 30, 460471. Parker, R., Sheth, U., 2007. P bodies and the control of mRNA translation and degradation. Mol. Cell 25, 635646. Petersen, C.P., Bordeleau, M.E., Pelletier, J., Sharp, P.A., 2006. Short RNAs repress translation after initiation in mammalian cells. Mol Cell 21, 533542. Pillai, R.S., Bhattacharyya, S.N., Artus, C.G., Zoller, T., Cougot, N., Basyuk, E., Bertrand, E., Filipowicz, W., 2005. Inhibition of translational initiation by Let-7 MicroRNA in human cells. Science 309, 15731676. Pillai, R.S., Bhattacharyya, S.N., Filipowicz, W., 2007. Repression of protein synthesis by miRNAs: how many mechanisms? Trends Cell Biol. 17, 118126. Reinhart, B.J., Bartel, D.P., 2002. Small RNAs correspond to centromere heterochromatic repeats. Science 297, 1831. Selbach, M., Schwanhausser, B., Thierfelder, N., Fang, Z., Khanin, R., Rajewsky, N., 2008. Widespread changes in protein synthesis induced by microRNAs. Nature 455, 5863. Tay, Y., Zhang, J., Thomson, A.M., Lim, B., Rigoutsos, I., 2008. MicroRNAs to Nanog, Oct4 and Sox2 coding regions modulate embryonic stem cell differentiation. Nature 455, 11241128. Thermann, R., Hentze, M.W., 2007. Drosophila miR2 induces pseudo-polysomes and inhibits translation initiation. Nature 447, 875878. Tsai, N.P., Lin, Y.L., Wei, L.N., 2009. Micro-RNA mir346 targets the 5UTR of RIP140 mRNA and up-regulates its protein expression. Biochem. J. 424, 411418. Vasudevan, S., Tong, Y., Steitz, J.A., 2007. Switching from repression to activation: microRNAs can up-regulate translation. Science 318, 19311934. Vella, M.C., Choi, E.Y., Lin, S.Y., Reinert, K., Slack, F.J., 2004a. The C. elegans microRNA let7 binds to imperfect let-7 complementary sites from the lin-41 3UTR. Genes Dev. 18, 132137. Vella, M.C., Reinert, K., Slack, F.J., 2004b. Architecture of a validated microRNA:target interaction. Chem. Biol. 11, 16191623. Vinther, J., Hedegaard, M.M., Gardner, P.P., Andersen, J.S., Arctander, P., 2006. Identication of miRNA targets with stable isotope labeling by amino acids in cell culture. Nucleic Acids Res. 34, e107. Ziegelbauer, J.M., Sullivan, C.S., Ganem, D., 2009. Tandem array-based expression screens identify host mRNA targets of virus-encoded microRNAs. Nat. Genet. 41, 130134.

Vous aimerez peut-être aussi