You are on page 1of 6

Theme Issue Article © Schattauer 2012 605

Regulation of microRNA biogenesis and function

MicroRNAs in vascular biology


Thomas Treiber; Nora Treiber; Gunter Meister
Biochemistry Center Regensburg (BZR), Laboratory for RNA Biology, University of Regensburg, Regensburg, Germany

Summary are typically located in the 3’ untranslated region (UTR) of mRNAs lead-
MicroRNAs (miRNAs) are considered as key regulators of literally all ing to inhibition of gene expression. MiRNA activity and abundance is
cellular pathways. Therefore, miRNA biosynthesis and their individual regulated on various levels ranging from transcription and processing
cellular functions must be tightly regulated as well. MiRNAs are tran- to target site binding and miRNA stability. Recent advances in our
scribed as primary transcripts, which are processed to mature miRNAs understanding of how miRNA activity is regulated in mammalian cells
in two consecutive maturation steps. Finally, the mature miRNA is incor- are summarised and discussed in this review article.
porated into a miRNA-protein complex, where it directly interacts with
a member of the Argonaute (Ago) protein family. The miRNA guides Keywords
such protein complexes to partial complementary target sites, which microRNAs, Dicer, Drosha, Argonaute, gene silencing

Correspondence to: Received: December 15, 2011


Gunter Meister Accepted after minor revision: January 11, 2012
Biochemistry Center Regensburg (BZR) Prepublished online: February 8, 2012
Laboratory for RNA Biology, University of Regensburg doi:10.1160/TH11-12-0836
Universitätsstrasse 31, 93053 Regensburg, Germany Thromb Haemost 2012; 107: 605–610
Tel.: +49 941 943 2847, Fax: +49 941 943 2936
E-mail: gunter.meister@vkl.uni-regensburg.de

Introduction periments in conjunction with mRNA level measurements reveal-


ed that mRNA decay accounts for approximately 85%, while trans-
MicroRNAs (miRNAs) are short non-coding RNAs that are viewed lational repression contributes only about 15% to miRNA-guided
as fundamental regulators of cell function (1, 2). MiRNAs are ap- gene silencing effects in mammals (8).
proximately 20–25 nucleotides (nt) long and are generated from MiRNA-guided gene silencing is mediated by Ago proteins,
double stranded RNA precursors (see below). One strand is se- which are the direct binding partners of mature miRNAs and
lected and interacts with a member of the Ago protein family to therefore central components of miRISC. In mammals, four differ-
form a miRNA-induced silencing complex (miRISC), also referred ent Ago proteins exist (Ago1-Ago4) and they are characterised by
to as micro-ribonucleoprotein (miRNP). The miRNA guides such PAZ (PIWI-Argonaute-Zwille), MID and PIWI (P-element-in-
complexes to partially complementary target RNAs. Most of the duced wimpy testes) domains. The PAZ domain anchors the 3’ end
target sites are located within the 3’ untranslated region (UTR) of of the miRNA, while the MID domain contains a highly specific
mRNAs. However, functional miRNA binding sites in the 5’ UTR binding pocket for the 5’ end of the miRNA (9). In addition, the
as well as in the open reading frame have also been reported (3, 4). MID domain discriminates between the four different bases at the
Nucleotides 2–8 of the miRNA are particularly important for pair- 5’ terminal nucleotide and prefers binding to uridines and, with a
ing with the target mRNA. This sequence motif is referred to as the slightly lower affinity, to adenosines explaining the U bias at the 5’
miRNA seed sequence (1). Depending on the recognition site, end of miRNAs (10). The PIWI domain is structurally similar to
binding of miRISC to the cognate target can have different out- RNase H and indeed some PIWI domains possess endonuclease
comes (5). In case the target site is perfectly complementary to the activity and mediate target RNA cleavage in RNAi (11, 12). In
miRNA, the miRNA functions like short interfering RNAs (siR- mammals, only Ago2 is endonucleolytically active (13, 14).
NAs) and the target is sequence-specifically cleaved by miRISC. MiRNA-guided regulation of gene expression has been impli-
This is very rare in mammals but more frequent in plants. Binding cated in literally every cellular pathway. In addition, each cell type
to partially complementary target sites is the rule in mammals and expresses a specific subset of miRNAs to ensure that cell type spe-
leads to repression of translation or degradation of the target tran- cific mRNA profiles are established and maintained. For example,
script (6). This degradation process, however, is different to RNAi- expression of a neuron-specific miRNA in non-neuronal cells re-
like mechanisms and involves the recruitment of deadenylase sults in shifting the global gene expression program towards tran-
complexes such as the CCR4-NOT complex to the mRNA to re- script profiles typically found in neurons (15). Another prominent
move or shorten the poly(A) tail. Poly(A) tail shortening induces example for the importance of miRNAs is the effect of a number of
the removal of the 5’ cap of the mRNA, a process referred to as de- miRNAs on the reprogramming of somatic cells to induced pluri-
capping. Uncapped mRNAs are rapidly removed from the cell by 5’ potent stem cells (iPS) (16).
to 3’ exoribonucleases such as Xrn1 (7). Ribosome profiling ex-

Thrombosis and Haemostasis 107.4/2012

Downloaded from www.thrombosis-online.com on 2016-08-02 | IP: 152.173.112.83


For personal or educational use only. No other uses without permission. All rights reserved.
606 Treiber et al. microRNA-guided gene silencing and its regulation

Given their fundamental cellular roles, it is not surprising that cessed by the RNase III enzyme Dicer (35). This enzyme binds the
miRNAs are affected in many diseases. Almost all types of cancer 3’ overhang of the pre-miRNA with its PAZ domain and thereby
MicroRNAs in vascular biology

have been analysed and miRNA missexpression was frequently positions the substrate correctly for the cleavage by the two cata-
noted (17, 18). Indeed, different cancers have different miRNA lytic domains 22 nt upstream within the double stranded stem (36,
profiles, which can even be found in the serum of patients (19). 37). A recent study has reported that in metazoa, the 5’ end of the
The functions of such circulating miRNAs have not been ident- pre-miRNA is contacted by Dicer proteins as well. This interaction
ified, but profiling of serum miRNAs might be a powerful ap- contributes to pre-miRNA recognition and correct processing
proach for early cancer diagnosis. (38). The result of Dicer cleavage is a double stranded RNA of 22 nt
Disease-causing changes in miRNA levels can have many differ- in length with 3’ overhangs of 2nt on both ends (1). One of the
ent reasons (6, 20). i) Similar to other genes, transcription can be strands (the mature miRNA) is transferred into an Ago protein,
positively or negatively affected. ii) miRNA processing can be regu- whereas the other strand (the star (*)-strand) is degraded. In
lated at each individual step (see below). iii) miRNA binding to mammals, the strand selection and RISC assembly is accomplished
Ago proteins as well as the activity of Ago proteins can be altered. by a complex that contains Dicer, Ago and the double stranded
iv) Accessibility of miRNA target sites on mRNAs can be changed. RNA binding protein TRBP (39–41). Statistical analyses have dis-
v) Other RNAs can function as sponges and sequester miRNAs covered that generally the strand with the less stable base pairing at
thereby regulating their activity (21). Recent progress towards the the 5’ end is chosen as guide strand (42, 43), which requires an
understanding of miRNA regulation will be summarised in several analysis of the thermodynamic properties of the double stranded
chapters of this review. miRNA after dicing. Recently, it has been proposed that the sensor
for the thermodynamic asymmetry is the helicase domain of
human Dicer itself. In this model, the double stranded Dicer cleav-
age product is repositioned on Dicer after the cleavage reaction.
miRNA biogenesis During this process, the helicase domain of Dicer senses the
thermodynamic stability of the ends and positions the double
MiRNAs are transcribed as parts of longer primary transcripts stranded RNA in an orientation that allows for correct guide
(pri-miRNAs) that are generated mainly by RNA polymerase II strand incorporation into RISC (씰Fig. 1) (44).
(22, 23). MiRNA genes are frequently located in transcripts of pro-
tein-coding genes, where they mainly reside in introns (24, 25).
Other miRNAs are transcribed as part of long non-coding RNAs
and are often arranged as clusters in the genome, which leads to Regulation of miRNA biogenesis
one pri-miRNA being subsequently processed into several func-
tional miRNAs (24). Like other Pol II transcripts, pri-miRNAs pos- Given the tremendous impact of miRNA-guided gene regulation
sess a 5’ cap and a 3’ poly-A-tail (23). Within the primary tran- on almost all aspects of cellular biology, it is not surprising that
scripts, miRNAs form stem-loop structures, which contain the ma- miRNA levels are tightly controlled and that deregulation can lead
ture miRNA as part of an imperfectly paired double stranded stem to various diseases including cancer (6, 17, 20).
connected by a short terminal loop. In the canonical miRNA bio- The first and one of the most important layers governing
genesis pathway, these structures are recognised by the micropro- miRNA abundance is the regulation of pri-miRNA transcription.
cessor complex, a multiprotein complex with the two core compo- The transcription of miRNAs encoded within introns of protein
nents Drosha and Di George Syndrome critical region gene 8 coding genes is mostly driven by the promoter of the host gene
(DGCR8) (26–29). The double-stranded RNA binding protein leading to a strong correlation of mRNA and miRNA expression.
DGCR8 binds to the base of the stem loop structure and thereby However, an analysis of chromatin signatures characteristic for
guides the positioning of the RNase III enzyme Drosha, which con- promoters has identified additional promoters for about one third
stitutes the catalytic center of the complex (30). Drosha cleaves the of the intronic miRNAs enabling a separate regulation from the
double-stranded stem about 11 bp from the base and generates a host gene (45). Intergenic miRNAs have dedicated promoters,
two nucleotide (nt) overhang at the 3’ end (27, 30). This cleavage which show all features commonly associated with Pol II-mediated
reaction liberates a hairpin-shaped RNA molecule of 70–100 bp transcription such as histone marks, CpG islands, transcription
called miRNA precursor or pre-miRNA. The mammalian micro- factor binding sites etc. (45). Clustered miRNAs share one pro-
processor contains several additional protein factors, most of moter and are coregulated as part of long pri-miRNAs. Via the pro-
which have not been assigned a specific function in miRNA pro- moters, pri-miRNA transcription can be strongly regulated by
cessing yet (27). Notably, the two DEAD-box RNA helicases DDX5 binding of transcription factors. The tumour suppressor p53, for
and DDX17 (also known as p68 and p72) have been identified as instance, has been shown to upregulate the transcription of
integral part of the microprocessor and are necessary for the effi- miR-34 family members, which in turn repress important factors
cient cleavage of a subset of pri-miRNAs (31). for cell proliferation and survival, such as Bcl2 and Cdk4 and 6
The pre-miRNAs are specifically recognised by the nuclear ex- (46–49).
port receptor Exportin 5 and exported in a Ran-GTP dependent In addition to the transcriptional regulation, it was noted that
manner (32–34). In the cytosol, the pre-miRNA is further pro- also the processing of pri-miRNA transcripts can be the limiting

Thrombosis and Haemostasis 107.4/2012 © Schattauer 2012

Downloaded from www.thrombosis-online.com on 2016-08-02 | IP: 152.173.112.83


For personal or educational use only. No other uses without permission. All rights reserved.
Treiber et al. microRNA-guided gene silencing and its regulation 607

factor for miRNA expression and can be regulated at different steps


leading to the accumulation of specific precursor forms (50, 51).

MicroRNAs in vascular biology


This posttranscriptional regulation can affect a large number of
miRNAs and is especially prominent in cancer samples and cell
lines (50, 52). Since these discoveries, a small but increasing
number of mechanisms that specifically regulate the processing of
miRNA subsets have been reported and will be discussed below.

Regulation of Drosha processing


An emerging group of proteins that can modulate pri-miRNA pro-
cessing by the microprocessor complex in response to diverse sti-
muli are transcription factors. For example, activation of Smad
proteins by stimulation of cells with bone morphogenic protein
(BMP) or tumour growth factor β (TGF-β) can stimulate the
maturation of specific miRNAs. This activity is independent of
Smad phosphorylation and binding of the Co-Smad4 and leads to
increased recruitment of the pri-miRNAs to the microprocessor
complex, enabling a more efficient cleavage by Drosha (53). Analy-
sis of the about 20 miRNAs that are affected by this regulation re-
vealed a consensus sequence that strongly resembles the Smad-
binding element in DNA and is located in the double stranded
stem of the pri-miRNAs. This motif is bound by the DNA-binding
MH1 domain of Smad1, 3 and 5 and establishes the specificity of
the regulated miRNAs (54). Another well-characterised Smad do-
main, the MH2 domain, binds to p68 (DDX5), which is part of the
microprocessor complex and is necessary for the induction of
miRNA processing by the Smad proteins (씰Fig. 1) (53).
Similarly, activation of the p53 pathway through DNA damage
leads to an association of the p53 protein with the microprocessor
complex via the p68 helicase (씰Fig. 1). This recruitment causes
increased processing of a set of pri-miRNAs including mir-145,
Figure 1: MiRNAs are transcribed as primary miRNA transcripts (pri-
which exerts a tumour suppressive function via repression of
miRNAs) in the nucleus of eukaryotic cells. In a first processing step, the
c-myc (55). In contrast to the Smad-regulated miRNAs, no com- microprocessor complex containing the catalytic subunit Drosha, its co-fac-
mon sequence motif could be delineated from the induced miR- tor DGCR8 and a number of other proteins including p68 and p72, liberates
NAs and it remains unclear how the target-miRNAs are selected. a hairpin-structured miRNA precursor (pre-miRNA). Pre-miRNAs are ex-
An alternative mechanism is used by the estrogen receptor ported to the cytoplasm by the export receptor Exportin 5. In the cytoplasm,
alpha (ERα), which associates with the microprocessor complex Dicer cleaves the pre-miRNA molecule and produces a short-lived double
and inhibits the cleavage of specific miRNAs after ligand binding stranded intermediate. One strand is selected and becomes the mature
(씰Fig. 1). Again, the helicase subunits of the microprocessor p68 miRNA whereas the other strand (referred to as miRNA-star) is degraded. The
and p72 are crucial for the recruitment of ERα (56). Interestingly, mature miRNA directly interacts with a member of the Argonaute protein
several estrogen-regulated miRNAs target mRNAs that are tran- family. MiRNA processing is regulated at many different steps. Factors that
scriptionally upregulated during estrogen-response. Thus, the in- positively influence miRNA processing or function are shown in green. Fac-
tors that inhibit miRNA maturation/function are highlighted in red.
hibition of miRNA maturation by ERα synergises with its tran-
scriptional effects by stabilising the target mRNAs (56). However,
how the specificity for the regulated miRNAs is achieved, needs to
be elucidated. stranded GGG-triplet. KHSRP binds to pri-miRNAs including
A second group of factors regulating microprocessor cleavage pri-let-7a and is necessary for microprocessor cleavage. Knock-
of a subset of miRNAs are RNA binding proteins that also have a down of KHSRP leads to a strong decrease in mature let-7a levels.
known role in RNA splicing (씰Fig. 1). The KH-type splicing regu- KHSRP is present in most cells and constitutively promotes pro-
latory protein (KHSRP) binds with high affinity to conserved ter- cessing of so far 14 identified miRNA targets (57). Its activity can
minal loop regions of a specific set of miRNAs. The binding motif be augmented by phosphorylation of three serine residues by the
is similar to binding sites in mRNA and consists of a single ATM kinase, which increases the affinity of KHSRP for the pri-

© Schattauer 2012 Thrombosis and Haemostasis 107.4/2012

Downloaded from www.thrombosis-online.com on 2016-08-02 | IP: 152.173.112.83


For personal or educational use only. No other uses without permission. All rights reserved.
608 Treiber et al. microRNA-guided gene silencing and its regulation

miRNA-loops in response to DNA damage (58). The stimulatory Regulation of Dicer processing
role of KHSRP in miRNA processing is antagonised by hnRNPA1,
MicroRNAs in vascular biology

which binds to the same site in pri-miRNAs and competes with The best-studied regulator of pre-miR processing by Dicer is the
KHSRP binding (59). In addition to its inhibitory role, hnRNPA1 protein Lin-28 (씰Fig. 1). In an evolutionary highly conserved
can also activate microprocessor cleavage of pri-miR-18a by bind- mechanism, this protein binds to the terminal loop of almost all
ing to the double stranded stem resulting in a more relaxed local let-7-family miRNAs (66–68). This high affinity interaction is me-
helix structure more favourable for Drosha cleavage (60, 61). Inter- diated by a cold shock domain and two zinc fingers that display a
estingly, in spite of being part of the large miR-17∼92 cluster, high homology to the NCp7 protein of HIV-1 (69–71). Lin-28 is
miR-18a is the only member that is regulated post-transcription- highly expressed in embryonic stem cells and leads to a complete
ally by hnRNPA1 (60, 61). This observation illustrates how regu- block in let-7 maturation thus ensuring that the expression of sev-
lation of miRNA processing is used to uncouple the expression lev- eral let-7 targets (including c-myc) that are essential for the main-
els of individual clustered miRNAs through posttranscriptional tenance of the pluripotent state, remain unrepressed. During dif-
mechanisms. ferentiation, the expression of Lin-28 is lost, and mature let-7 miR-
Similar to the regulation of miR-18 by hnRNPA1, the SR-pro- NAs can be produced (66). Repression of Dicer processing involves
tein SF2/ASF can bind to the lower stem of pri-miR-7 and promote the cytoplasmic polyuridylation of the pre-miRNA at its 3’ end
Drosha cleavage. As the translation of SF2/ASF mRNA is repressed (72), a reaction catalysed by terminal uridyl transferases (TUT-
by miR-7, this leads to the formation of a negative feedback loop ases). Recent high-throughput sequencing data of miRNAs has
that attenuates SF2 expression (62). shown that a considerable amount of pre-miRNAs carry at least a
Adenosine deaminase acting on RNA (ADAR) enzymes bind single U added to their 3’ end (73). Pre-let-7 bound Lin-28 recruits
double stranded RNA and deaminate adenosine to inosine. Apart a specific TUTase, TUT4, and thereby acts as a processivity-factor
from their function in mRNA editing they can also accept certain mediating the specific polyuridylation of its bound pre-miRNA
miRNA precursors as substrates resulting in an I-U wobble base (74–76). In addition to the regulation of pre-miRNA processing by
pair in the modified RNA. For miR-142 it has been shown that Dicer, it has been proposed that Lin-28 also plays a role in the nu-
Drosha processing of the edited pri-miRNA is substantially im- cleus by blocking Drosha cleavage. This has been shown in Caenor-
paired. Instead, the pri-miRNA is recognised and degraded by the habditis elegans where Lin-28 binds pri-let-7 co-transcriptionally
I-U specific nuclease Tudor-SN, resulting in a severe decrease in and inhibits microprocessor activity (77). Very recently, it has been
mature miRNA levels (63). reported in mammals, that a homolog of Lin-28, Lin-28b, re-
presses Drosha processing by sequestering pri-let-7 to nucleoli,
where they are not accessible for the microprocessor complex (78).
Apart from stem cells, Lin-28 and Lin-28b are expressed in special-
Regulation of pre-miRNA export ised tissues and are frequently upregulated in cancer, where they
promote tumour growth by repressing let-7 miRNAs.
The pre-miRNA export receptor Exportin 5 is, like other importin- Members of the let-7 family are not the only targets of repres-
β/karyopherin family proteins, composed of HEAT repeats that sion by Lin-28 and Tut4. It has been shown that in cardiac tissue,
form a solenoid-shaped molecule with a large binding groove for pre-miR-1 can be uridylated and degraded in a Lin-28-dependent
the cargo. In Exportin 5, this binding region is strongly basic and manner. Yet, under normal conditions, this process is prevented by
can perfectly accomodate the negatively charged RNA double helix the binding of the MBNL1-protein to the terminal loop of miR-1,
of the pre-miRNA stem. An additional small pocket at the base of thereby competing with Lin-28 for binding. In myotonic dys-
the groove binds the 2 nt 3’ overhang and ensures the specificity for trophy, however, MBNL1 is sequestered by binding to expanded
Drosha cleavage products. The terminal loop is not bound and can CUG or CCUG sequences that cause the disease. Thus, Lin-28 can
vary in size and shape (64). This mode of binding might allow for bind pre-miR-1 and the expression of the mature miRNA is lost,
the co-export of proteins bound to the terminal loop but not the which contributes to the cardiac manifestation of the dystrophy
double stranded pre-miRNA stem. through dysregulation of membrane channels (79).
C-terminally truncated mutations of Exportin 5 have been
identified in tumours with microsatellite instability and result in a
global defect of pre-miRNA export. The resulting general decrease
of mature miRNA levels is a hallmark of miRNA profiles of cancer Regulation of miRISC stability
cells (65). So far there is only indirect evidence for specific regu-
lation of nuclear export of individual miRNA precursors. Several The final regulatory level of miRNA abundance is the stability of
pri-miRNAs including mir-31 are processed by the microproces- miRISC. More and more specific mechanisms that can destabilise
sor but retained in the nucleus in most cell lines (52). However, the miRNAs as well as miRISC protein components are uncovered. In
detailed mechanisms as well as the functional consequences re- stem cells, the tripartite motif protein Lin-41 acts as specific ubi-
main to be elucidated. quitin ligase for Ago proteins. As Ago proteins appear to be limiting
for the activity of the miRNA pathway, this results in the global at-
tenuation of miRNA-guided gene silencing (80). Upon differenti-

Thrombosis and Haemostasis 107.4/2012 © Schattauer 2012

Downloaded from www.thrombosis-online.com on 2016-08-02 | IP: 152.173.112.83


For personal or educational use only. No other uses without permission. All rights reserved.
Treiber et al. microRNA-guided gene silencing and its regulation 609

ation of stem cells, Lin-41 expression is downregulated, at least References


partially as result of repression by let-7 family members that are re-

MicroRNAs in vascular biology


1. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 2009;
leased from the maturation block imposed by Lin-28 and destabi- 136: 215–233.
lise Lin-41 mRNA (80). 2. Carthew RW, Sontheimer EJ. Origins and Mechanisms of miRNAs and siRNAs.
On the miRNA side, it is becoming evident that different miR- Cell 2009; 136: 642–655.
NAs have distinct inherent half-lives that might even be encoded in 3. Orom UA, Nielsen FC, Lund AH. MicroRNA-10a binds the 5'UTR of ribosomal
protein mRNAs and enhances their translation. Mol Cell 2008; 30: 460–471.
their sequence. miR-382, for example, is relatively short lived and 4. Tay Y, Zhang J, Thomson AM, et al. MicroRNAs to Nanog, Oct4 and Sox2 coding
degraded by the exosome. A sequence in the 3’ end of the miRNA regions modulate embryonic stem cell differentiation. Nature 2008; 455:
is crucial in this process (81). Moreover, several retinal miRNAs are 1124–1128.
5. Pillai RS, Bhattacharyya SN, Filipowicz W. Repression of protein synthesis by
rapidly turned over upon light stimulation (82). The detailed
miRNAs: how many mechanisms? Trends Cell Biol 2007; 17: 118–126.
mechanisms of active miRNA turn over in mammals, however, has 6. Krol J, Loedige I, Filipowicz W. The widespread regulation of microRNA biogen-
not been elucidated. In C. elegans, it has been demonstrated that esis, function and decay. Nat Rev Genet 2010; 11: 597–610.
miRNAs, which are not bound to target RNAs, are degraded by the 7. Huntzinger E, Izaurralde E. Gene silencing by microRNAs: contributions of
translational repression and mRNA decay. Nat Rev Genet 2011; 12: 99–110.
5’ to 3’ exoribonuclease Xrn2 (83). It is therefore very likely that 8. Guo H, Ingolia NT, Weissman JS, et al. Mammalian microRNAs predominantly
more, so far unrecognised factors might influence miRNA stability act to decrease target mRNA levels. Nature 2010; 466: 835–840.
and thereby regulate gene silencing. 9. Jinek M, Doudna JA. A three-dimensional view of the molecular machinery of
RNA interference. Nature 2009; 457: 405–412.
10. Frank F, Sonenberg N, Nagar B. Structural basis for 5'-nucleotide base-specific
recognition of guide RNA by human AGO2. Nature 2010; 465: 818–822.
11. Yuan YR, Pei Y, Ma JB, et al. Crystal Structure of A. aeolicus Argonaute, a Site-Spe-
Outlook cific DNA-Guided Endoribonuclease, Provides Insights into RISC-Mediated
mRNA Cleavage. Mol Cell 2005; 19: 405–419.
12. Song JJ, Smith SK, Hannon GJ, et al. Crystal structure of Argonaute and its impli-
It is becoming more and more apparent that miRNA activity is cations for RISC slicer activity. Science 2004; 305: 1434–1437.
heavily regulated at all steps of the miRNA pathway. It is likely that 13. Liu J, Carmell MA, Rivas FV, et al. Argonaute2 is the catalytic engine of mamma-
numerous, so far not identified RNA binding proteins are engaged lian RNAi. Science 2004; 305: 1437–1441.
in the regulation of miRNA function. It might even be possible that 14. Meister G, Landthaler M, Patkaniowska A, et al. Human Argonaute2 Mediates
RNA Cleavage Targeted by miRNAs and siRNAs. Mol Cell 2004; 15: 185–197.
each miRNA family or miRNA cluster possesses its own repertoire 15. Lim LP, Lau NC, Garrett-Engele P, et al. Microarray analysis shows that some
of regulatory proteins. In addition, key protein components of the microRNAs downregulate large numbers of target mRNAs. Nature 2005; 433:
miRNA pathway are subject to phosphorylation (84). Currently, 769–773.
16. Mallanna SK, Rizzino A. Emerging roles of microRNAs in the control of em-
we do not understand how miRNA pathways are embedded into
bryonic stem cells and the generation of induced pluripotent stem cells. Dev Biol
cellular signalling networks and it is possible that kinases and 2010; 344: 16–25.
phosphatases are involved in the interplay of miRNAs and signal- 17. Croce CM. Causes and consequences of microRNA dysregulation in cancer. Nat
ling molecules. Rev Genet 2009; 10: 704–714.
18. Farazi TA, Horlings HM, Ten Hoeve JJ, et al. MicroRNA sequence and expression
Very recently, pseudogenes have been implicated in the regu- analysis in breast tumors by deep sequencing. Cancer Res 2011; 71: 4443–4453.
lation of miRNA activity as well. Numerous conserved pseudo- 19. Wittmann J, Jack HM. Serum microRNAs as powerful cancer biomarkers. Bio-
genes exist in the human genome and many of them contain 3’ chim Biophys Acta 2010; 1806: 200–207.
UTRs with conserved miRNA binding sites. Interestingly, these 20. Winter J, Jung S, Keller S, et al. Many roads to maturity: microRNA biogenesis
pathways and their regulation. Nat Cell Biol 2009; 11: 228–234.
binding sites are active and function as decoys or sponges seques- 21. Salmena L, Poliseno L, Tay Y, et al. A ceRNA hypothesis: the Rosetta Stone of a
tering miRNAs and preventing them from binding to their cognate hidden RNA language? Cell 2011; 146: 353–358.
mRNA targets. Such RNAs have been termed competing endogen- 22. Lee Y, Kim M, Han J, et al. MicroRNA genes are transcribed by RNA polymerase
ous RNAs (ceRNAs) (85). These new findings highlight the com- II. Embo J 2004; 23: 4051–4060.
23. Cai X, Hagedorn CH, Cullen BR. Human microRNAs are processed from capped,
plexity of regulatory miRNA networks. In order to understand the polyadenylated transcripts that can also function as mRNAs. RNA 2004; 10:
impact of miRNAs on disease and the development of miRNA- 1957–1966.
based therapeutics, it is essential to characterise such miRNA net- 24. Rodriguez A, Griffiths-Jones S, Ashurst JL, et al. Identification of mammalian
works in molecular detail. microRNA host genes and transcription units. Genome Res 2004; 14: 1902–1910.
25. Smalheiser NR. EST analyses predict the existence of a population of chimeric
microRNA precursor-mRNA transcripts expressed in normal human and mouse
Acknowledgements tissues. Genome Biol 2003; 4: 403.
We apologise to those, whose work was not cited due to space con- 26. Lee Y, Ahn C, Han J, et al. The nuclear RNase III Drosha initiates microRNA pro-
straints. Our research is supported in part by grants of the Euro- cessing. Nature 2003; 425: 415–419.
27. Gregory RI, Yan KP, Amuthan G, et al. The Microprocessor complex mediates the
pean Research Council (ERC, sRNAs to G.M.), the EU FP7 project genesis of microRNAs. Nature 2004; 432: 235–240.
‘ONCOMIRs’, the Bavarian Genome Research Network (BayGene 28. Denli AM, Tops BB, Plasterk RH, et al. Processing of primary microRNAs by the
to G.M.) and the Deutsche Forschungsgemeinschaft (DFG). Microprocessor complex. Nature 2004; 432: 231–235.
29. Landthaler M, Yalcin A, Tuschl T. The human DiGeorge syndrome critical region
gene 8 and Its D. melanogaster homolog are required for miRNA biogenesis. Curr
Conflicts of interest Biol 2004; 14: 2162–2167.
None declared. 30. Han J, Lee Y, Yeom KH, et al. Molecular basis for the recognition of primary
microRNAs by the Drosha-DGCR8 complex. Cell 2006; 125: 887–901.

© Schattauer 2012 Thrombosis and Haemostasis 107.4/2012

Downloaded from www.thrombosis-online.com on 2016-08-02 | IP: 152.173.112.83


For personal or educational use only. No other uses without permission. All rights reserved.
610 Treiber et al. microRNA-guided gene silencing and its regulation

31. Fukuda T, Yamagata K, Fujiyama S, et al. DEAD-box RNA helicase subunits of the 60. Michlewski G, Guil S, Semple CA, et al. Posttranscriptional regulation of miRNAs
Drosha complex are required for processing of rRNA and a subset of microRNAs. harboring conserved terminal loops. Mol Cell 2008; 32: 383–393.
MicroRNAs in vascular biology

Nat Cell Biol 2007; 9: 604–611. 61. Guil S, Caceres JF. The multifunctional RNA-binding protein hnRNP A1 is
32. Lund E, Guttinger S, Calado A, et al. Nuclear export of microRNA precursors. required for processing of miR-18a. Nat Struct Mol Biol 2007; 14: 591–596.
Science 2004; 303: 95–98. 62. Wu H, Sun S, Tu K, et al. A splicing-independent function of SF2/ASF in microR-
33. Yi R, Qin Y, Macara IG, et al. Exportin-5 mediates the nuclear export of pre- NA processing. Mol Cell 2010; 38: 67–77.
microRNAs and short hairpin RNAs. Genes Dev 2003; 17: 3011–3016. 63. Yang W, Chendrimada TP, Wang Q, et al. Modulation of microRNA processing
34. Bohnsack MT, Czaplinski K, Gorlich D. Exportin 5 is a RanGTP-dependent and expression through RNA editing by ADAR deaminases. Nat Struct Mol Biol
dsRNA-binding protein that mediates nuclear export of pre-miRNAs. RNA 2004; 2006; 13: 13–21.
10: 185–191. 64. Okada C, Yamashita E, Lee SJ, et al. A high-resolution structure of the pre-microR-
35. Grishok A, Pasquinelli AE, Conte D, et al. Genes and mechanisms related to RNA NA nuclear export machinery. Science 2009; 326: 1275–1279.
interference regulate expression of the small temporal RNAs that control C. el- 65. Melo SA, Moutinho C, Ropero S, et al. A genetic defect in exportin-5 traps precur-
egans developmental timing. Cell 2001; 106: 23–34. sor microRNAs in the nucleus of cancer cells. Cancer Cell 2010; 18: 303–315.
36. Macrae IJ, Zhou K, Li F, et al. Structural basis for double-stranded RNA process- 66. Viswanathan SR, Daley GQ, Gregory RI. Selective blockade of microRNA pro-
ing by Dicer. Science 2006; 311: 195–198. cessing by Lin28. Science 2008; 320: 97–100.
37. Zhang H, Kolb FA, Jaskiewicz L, et al. Single processing center models for human 67. Rybak A, Fuchs H, Smirnova L, et al. A feedback loop comprising lin-28 and let-7
Dicer and bacterial RNase III. Cell 2004; 118: 57–68. controls pre-let-7 maturation during neural stem-cell commitment. Nat Cell Biol
38. Park JE, Heo I, Tian Y, et al. Dicer recognizes the 5' end of RNA for efficient and ac- 2008; 10: 987–993.
curate processing. Nature 2011; 475: 201–205. 68. Newman MA, Thomson JM, Hammond SM. Lin-28 interaction with the Let-7
39. Chendrimada TP, Gregory RI, Kumaraswamy E, et al. TRBP recruits the Dicer precursor loop mediates regulated microRNA processing. RNA 2008; 14:
complex to Ago2 for microRNA processing and gene silencing. Nature 2005; 436: 1539–1549.
740–744. 69. Piskounova E, Viswanathan SR, Janas M, et al. Determinants of microRNA pro-
40. Haase AD, Jaskiewicz L, Zhang H, et al. TRBP, a regulator of cellular PKR and cessing inhibition by the developmentally regulated RNA-binding protein Lin28.
HIV-1 virus expression, interacts with Dicer and functions in RNA silencing. J Biol Chem 2008; 283: 21310–21314.
EMBO Rep 2005; 6: 961–967. 70. Desjardins A, Yang A, Bouvette J, et al. Importance of the NCp7-like domain in the
41. Gregory RI, Chendrimada TP, Cooch N, et al. Human RISC couples microRNA recognition of pre-let-7g by the pluripotency factor Lin28. Nucleic Acids Res
biogenesis and posttranscriptional gene silencing. Cell 2005; 123: 631–640. 2011; epub ahead of print.
42. Khvorova A, Reynolds A, Jayasena SD. Functional siRNAs and miRNAs exhibit 71. Nam Y, Chen C, Gregory RI, et al. Molecular Basis for Interaction of let-7 MicroR-
strand bias. Cell 2003; 115: 209–216. NAs with Lin28. Cell 2011; 147: 1080–1091.
43. Schwarz DS, Hutvágner G, Du T, et al. Asymmetry in the assembly of the RNAi 72. Heo I, Joo C, Cho J, et al. Lin28 mediates the terminal uridylation of let-7 precur-
enzyme complex. Cell 2003; 115: 199–208. sor MicroRNA. Mol Cell 2008; 32: 276–284.
44. Noland CL, Ma E, Doudna JA. siRNA repositioning for guide strand selection by 73. Newman MA, Mani V, Hammond SM. Deep sequencing of microRNA precursors
human Dicer complexes. Mol Cell 2011; 43: 110–121. reveals extensive 3' end modification. RNA 2011; 17: 1795–1803.
45. Ozsolak F, Poling LL, Wang Z, et al. Chromatin structure analyses identify miRNA 74. Heo I, Joo C, Kim YK, et al. TUT4 in concert with Lin28 suppresses microRNA
promoters. Genes Dev 2008; 22: 3172–3183. biogenesis through pre-microRNA uridylation. Cell 2009; 138: 696–708.
46. Raver-Shapira N, Marciano E, Meiri E, et al. Transcriptional activation of miR-34a 75. Hagan JP, Piskounova E, Gregory RI. Lin28 recruits the TUTase Zcchc11 to inhibit
contributes to p53-mediated apoptosis. Mol Cell 2007; 26: 731–743. let-7 maturation in mouse embryonic stem cells. Nat Struct Mol Biol 2009; 16:
47. He L, He X, Lim LP, et al. A microRNA component of the p53 tumour suppressor 1021–1025.
network. Nature 2007; 447: 1130–1134. 76. Yeom KH, Heo I, Lee J, et al. Single-molecule approach to immunoprecipitated
48. Tarasov V, Jung P, Verdoodt B, et al. Differential regulation of microRNAs by p53 protein complexes: insights into miRNA uridylation. EMBO Rep 2011; 12:
revealed by massively parallel sequencing: miR-34a is a p53 target that induces 690–696.
apoptosis and G1-arrest. Cell Cycle 2007; 6: 1586–1593. 77. Van Wynsberghe PM, Kai ZS, Massirer KB, et al. LIN-28 co-transcriptionally
49. Chang TC, Wentzel EA, Kent OA, et al. Transactivation of miR-34a by p53 broadly binds primary let-7 to regulate miRNA maturation in Caenorhabditis elegans.
influences gene expression and promotes apoptosis. Mol Cell 2007; 26: 745–752. Nat Struct Mol Biol 2011; 18: 302–308.
50. Thomson JM, Newman M, Parker JS, et al. Extensive post-transcriptional regu- 78. Piskounova E, Polytarchou C, Thornton JE, et al. Lin28A and Lin28B Inhibit let-7
lation of microRNAs and its implications for cancer. Genes Dev 2006; 20: MicroRNA Biogenesis by Distinct Mechanisms. Cell 2011; 147: 1066–1079.
2202–2207. 79. Rau F, Freyermuth F, Fugier C, et al. Misregulation of miR-1 processing is associ-
51. Obernosterer G, Leuschner PJ, Alenius M, et al. Post-transcriptional regulation of ated with heart defects in myotonic dystrophy. Nat Struct Mol Biol 2011; 18:
microRNA expression. Rna 2006; 12: 1161–1167. 840–845.
52. Lee EJ, Baek M, Gusev Y, et al. Systematic evaluation of microRNA processing pat- 80. Rybak A, Fuchs H, Hadian K, et al. The let-7 target gene mouse lin-41 is a stem cell
terns in tissues, cell lines, and tumors. RNA 2008; 14: 35–42. specific E3 ubiquitin ligase for the miRNA pathway protein Ago2. Nat Cell Biol
53. Davis BN, Hilyard AC, Lagna G, et al. SMAD proteins control DROSHA-mediated 2009; 11: 1411–1420.
microRNA maturation. Nature 2008; 454: 56–61. 81. Bail S, Swerdel M, Liu H, et al. Differential regulation of microRNA stability. RNA
54. Davis BN, Hilyard AC, Nguyen PH, et al. Smad proteins bind a conserved RNA se- 2010; 16: 1032–1039.
quence to promote microRNA maturation by Drosha. Mol Cell 2010; 39: 82. Krol J, Busskamp V, Markiewicz I, et al. Characterizing light-regulated retinal
373–384. microRNAs reveals rapid turnover as a common property of neuronal microR-
55. Suzuki HI, Yamagata K, Sugimoto K, et al. Modulation of microRNA processing NAs. Cell 2010; 141: 618–631.
by p53. Nature 2009; 460: 529–533. 83. Chatterjee S, Grosshans H. Active turnover modulates mature microRNA activity
56. Yamagata K, Fujiyama S, Ito S, et al. Maturation of microRNA is hormonally regu- in Caenorhabditis elegans. Nature 2009; 461: 546–549.
lated by a nuclear receptor. Mol Cell 2009; 36: 340–347. 84. Rudel S, Meister G. Phosphorylation of Argonaute proteins: regulating gene regu-
57. Trabucchi M, Briata P, Garcia-Mayoral M, et al. The RNA-binding protein KSRP lators. Biochem J 2008; 413: e7–9.
promotes the biogenesis of a subset of microRNAs. Nature 2009; 459: 1010–1014. 85. Tay Y, Kats L, Salmena L, et al. Coding-Independent Regulation of the Tumor Sup-
58. Zhang X, Wan G, Berger FG, et al. The ATM kinase induces microRNA biogenesis pressor PTEN by Competing Endogenous mRNAs. Cell 2011; 147: 344–357.
in the DNA damage response. Mol Cell 2011; 41: 371–383.
59. Michlewski G, Caceres JF. Antagonistic role of hnRNP A1 and KSRP in the regu-
lation of let-7a biogenesis. Nat Struct Mol Biol 2010; 17: 1011–1018.

Thrombosis and Haemostasis 107.4/2012 © Schattauer 2012

Downloaded from www.thrombosis-online.com on 2016-08-02 | IP: 152.173.112.83


For personal or educational use only. No other uses without permission. All rights reserved.

You might also like