You are on page 1of 27

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/261707675

Micro RNA- Biogenesis, Mechanism of Action and Applications-A Review

Article · November 2011

CITATION READS
1 900

1 author:

Subhabrata Ghosh
University of Kalyani
14 PUBLICATIONS   106 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

DNA Fingerprinting View project

All content following this page was uploaded by Subhabrata Ghosh on 19 April 2014.

The user has requested enhancement of the downloaded file.


Available online at http://www.urpjournals.com

International Journal of Research in Biotechnology and Biochemistry

Universal Research Publications. All rights reserved

Review article
Micro RNA- Biogenesis, Mechanism of Action and Applications-A Review
Subhabrata Ghosh
Bose Institute, Kolkata-700009, India
Ph-+919903221547
Email-subhabrata09@yahoo.com
subhabrata.boseinstitute@gmail.com
Received 01 November 2011; accepted 14 November 2011
Abstract
MicroRNAs (miRNAs) are small RNA molecules that regulate gene expression post-transcriptionally. After the discovery of
the first miRNA in the roundworm Caenorhabditis elegans, these short regulatory RNAs have been found to be an abundant
class of RNAs in plants, animals, and DNA viruses. The discoveries of RNA interference (RNAi), known as remarkable
process where specific genes are silenced by small noncoding RNAs, have revolutionized our understanding of gene regulation
in plants and other organisms. Recently, Micro RNA is used as a tool for reduction of expression of several genes or to control
de expression of specific genes in eukaryotic genome. The aim of this review is to describe the biology, process of biogenesis
and mechanism of action of Micro RNA and also to underline the most contribution in the ―world of Micro RNA‖ from
biological mechanism to several applications including cancer treatment, neurodegenerative diseases and viral infections like
AIDS, Hepatitis, Swine Flu etc.
© 2011 Universal Research Publications. All rights reserved
Keywords: Micro RNA, Gene regulation, Cancer, Neurodegenarative disease, AIDS

1. Introduction: partially complementary to 7 conserved sites located in the


According to the current convention, a miRNA is defined as a 3′-untranslated region (UTR) of the lin-14 gene (Figure-1)21,
22
ssRNA of ~22 nucleotides in length, which is generated by . lin-14 encodes a nuclear protein, down-regulation of
the RNase-III-type enzyme Dicer from an endogenous which at the end of the first larval stage initiates the
transcript that contains a local hairpin structure1. developmental progression into the second larval
stage,21,23.The negative regulation of LIN-14 protein
1.1 Discovery of MicroRNA: expression requires an intact 3′ UTR of its mRNA14, as well
The founding member of the miRNA family, lin-4, was as a functional lin-4gene13. These genetic interactions
identified in C. elegans through a genetic screen for defects in inspired a series of molecular and biochemical studies
the temporal control of post-embryonic development16,17.In demonstrating that the direct, but imprecise, base pairing
C. elegans, cell lineages have distinct characteristics during 4 between lin-4 and the lin-14 3′ UTR was essential for the
different larval stages (L1–L4).Mutations in lin-4 disrupt the ability of lin-4 to control LIN-14 expression through the
temporal regulation of larval development, causing L1 (the regulation of protein synthesis24-26. Through an analogous
first larval stage)-specific cell-division patterns to reiterate at mechanism, lin-4 also negatively regulates the translation of
later developmental stages18.Opposite developmental lin-28, a cold-shock-domain protein that initiates the
phenotypes — omission of the L1 cell fates and premature developmental transition between the L2 and L3 stages27.
development into the L2 stage — are observed in worms that Compared with lin-14, lin-28 has fewer lin-4 binding sites,
are deficient for lin-1419. Even before the molecular which might lead to its translational repression being delayed
identification of lin-4 and lin-14, these loci were placed in the following lin-4 expression owing to less efficient lin-4
same regulatory pathway on the basis of their opposing binding28,27.The discovery of lin-4 and its target-specific
phenotypes and antagonistic genetic interactions20.Most translational inhibition hinted at a new mechanism of gene
genes identified from mutagenesis screens are protein-coding, regulation during development. In 2000, almost 7 years after
but lin-4 encodes a 22-nucleotide non-coding RNA that is the initial identification of lin-4, the second miRNA, let-7,
International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
11
was discovered, also using forward genetics in worms.let-7
encodes a temporally regulated 21-nucleotide small RNA that
controls the developmental transition from the L4 stage into
the adult stage 29-31. Similar to lin-4, let-7 performs its
function by binding to the 3′ UTR of lin-41and hbl-1 (lin-57),
and inhibiting their translation 29-33. The identification of let-7
not only provided another vivid example of developmental
regulation by small RNAs, but also raised the possibility that
such RNAs might be present in species other than nematodes.
Unlike lin-4, the orthologues of which in flies and mammals
initially escaped bio-informatic searches, and were only
recognized recently34,35,both let-7 and lin-41 are
evolutionarily conserved throughout metazoans, with
homologues that were readily detected in molluscs, sea
urchins, flies, mice and humans36.

1.2 Types and Distribution of small RNAs in Eukaryotic


system:
Apart from microRNAs (miRNAs), several other groups of
Figure 1. The molecular hallmarks of lin-4, the founding small RNAs have been described2: small interfering RNAs
member of the microRNA family. (siRNAs), tiny non-coding RNAs (tncRNAs) and a small
(a) The precursor structure and mature microRNA (miRNA)
modulatory RNA (smRNA). siRNAs are defined as 21–28-
sequence of lin-4 (b) Sequence complementarity between lin-4 (red) nucleotide RNAs, which are produced by Dicer from long
and the 3′-untranslated region (UTR) of lin-14 mRNA (blue).lin-4 double-stranded RNAs (dsRNAs). Small RNAs used in
is partially complementary to 7 sites in the lin-14 3′ UTR; its RNA-interference-mediated experiments are traditionally
binding to these sites of complementarity brings about repression of referred to as siRNAs, regardless of their origin. Endogenous
LIN-14 protein synthesis.[RISC- RNA-induced silencing complex]. siRNAs have also been discovered in various organisms.
Endogenous siRNAs can be further categorized into at least
three distinct sub-classes: trans-acting siRNAs (tasiRNAs),
repeat-associated siRNAs (rasiRNAs) and small-scan RNAs
(scnRNAs). tasiRNAs found in plants are ~21-nucleotide
RNAs that are encoded in intergenic regions, and they act on
mRNAs intrans to induce mRNA cleavage3,4,5. Some
endogenous siRNAs match repetitive elements in the sense or
antisense orientation and are therefore known as rasiRNAs6,7.
Evidence from Schizosaccharomyces pombe and Arabidopsis
thaliana indicates that rasiRNAs might function in
transcriptional gene silencing through methylation of histones
and/or DNA. siRNAs of ~28 nucleotides found in
Tetrahymena thermophila are known as scnRNA because
they might scan for regions in the DNA for genome
rearrangement8-10. The Argonaute family protein TWI1
mediates histone methylation, which is, in turn, required for
DNA elimination8. There are additional small RNAs of which
the biogenesis and function remain less clear, and which
hinders their classification. tncRNAs described in nematode
Figure 2. Genomic organization and structure of miRNA worms are similar to tasiRNAs11, although their origin from
genes. the long dsRNA precursor needs to be verified. A smRNA of
(a) Intronic miRNA in a protein-coding transcriptional unit (TU). As ~20 nucleotides that has been cloned from neural stem cells
an example, miR-10 in HOX4B gene is shown. The green triangle modulates transcription possibly by interacting with a
indicates the location of a miRNA stem-loop and the exons are transcription factor 12. The biogenesis of smRNAs is as yet
shown in yellow. unknown.
(b) Intronic miRNAs in a noncoding transcript. The miR-15a-16-1 Since the discovery of RNA interference (RNAi),
cluster is shown, which is found in the fourth intron of a previously efforts to identify endogenous small RNAs have led to the
defined noncoding RNA gene, DLEU2. discovery of hundreds of miRNAs in nematodes, fruit flies
(c) The structure of exonic miRNA in noncoding transcripts, such as
and humans. More than 500 different miRNAs have been
miR-155.

International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36


12
Figure 3. Possible mechanisms of actions for Drosha and Dicer.
The ‗single processing centre‘ model applies to both Drosha and Dicer. According to this model, one processing centre is formed in the
interface between two RNase III domains (RIIIDa and b).The processing centre contains two closely located catalytic sites that cleave two
nearby phosphodiester bonds on opposite RNA strands. The catalytic sites are composed of residues that correspond to E40, D44, D107 and
E110 of Aquifex aeolicus RNase III. This ‗single processing centre model‘ differs from the previous model, which assumed the existence of
two processing centres with the second catalytic site being composed of residues E37 and E64 in A. aeolicus RNase III. The double-stranded
RNA (dsRNA) substrate is laid on the cleft between RIIIDa (light orange) and RIIIDb (dark orange). The catalytic cleaves the 5′ strand.
(a) Drosha binds to a primary transcript (pri-miRNA) and introduces a cut at approximately two helical turns, ~22 nucleotides (nt site on the
RIIIDa side cleaves the 3′ strand, whereas another catalytic site on the RIIIDb), from the terminal loop. For simplicity, the domains of Drosha
other than the RIIIDs are not shown. It was proposed that DGCR8/Pasha might help the binding of the complex to RNA and/or orienting the
complex on pri-miRNA.
(b) Dicer cleaves at approximately two helical turns (~22 nucleotides) from the 3′ terminus. The PAZ domain of Dicer is believed to hold the
3′ end of pre-miRNA. The domains of Dicer other than the RIIIDs and the PAZ domain are not shown.

identified in animals and plants, where the number of miRNA cluster are often related to each other, suggesting that the
genes is expected to increase to 500–1000 per species, which gene cluster is a result of gene duplication. A miRNA gene
would comprise w2–3% of protein-coding genes13.At the cluster also often contains unrelated miRNAs. A plausible but
time of writing, the miRNA database (http://www.sanger yet-to-be validated possibility is that the clustered miRNAs
.ac.uk/Software/Rfam/mirna/)contained 114 Caenorhabditis are functionally related by virtue of targeting the same gene
elegans miRNAs, 78 Drosophila melanogaster miRNAs, 369 or different genes in the same pathway. It was initially
Danio rerio miRNAs, 122 Gallus gallus miRNAs, 326 Homo thought that most miRNA genes were located in intergenic
sapiens miRNAs and 117 Arabidopsis thaliana regions. However, recent analyses of miRNA gene locations
miRNAs14.Nearly all miRNAs are conserved in closely showed that the majority (w70%) of mammalian miRNA
related species and many have homologs in distant species. genes (161 out of 232) are located in defined transcription
At least a third of C. elegans miRNAs have homologs in units (TU)41. By combining up-to-date genome assemblies
humans15, suggesting that their functions could also be and expressed sequence tag (EST) databases, Rodriguez et al.
conserved throughout the evolution of animal lineages. Large demonstrated that many miRNA genes (117 out of 161) were
DNA viruses have also been found to carry miRNA genes: found in the introns in the sense orientation, which is more
five in Epstein–Barr virus, 12 in Kaposi sarcoma-associated than previously expected 41. Of these 117 intronic miRNAs,
herpesvirus, nine in mouse g-herpesvirus, and nine in human 90 miRNAs are in the introns of protein-coding genes,
cytomegalovirus. whereas 27 miRNAs are in the introns of noncoding RNAs
(ncRNAs). In some cases (14 miRNAs), miRNAs are present
1.3 Genomic Distribution and gene structure of Micro in either an exon or an intron (‗mixed‘) depending on the
RNA: alternative splicing pattern. So, miRNA genes can be
miRNA genes are scattered in all chromosomes in humans categorized based on their genomic locations: intronic
except for the Y chromosome. Approximately 50% of known miRNA in protein coding TU; intronic miRNA in noncoding
miRNAs are found in clusters37, 38, 39 and they are transcribed TU; and exonic miRNA in noncoding TU (Figure-2).
as polycistronic primary transcripts40.The miRNAs in a given ‗Mixed‘ miRNA genes can be assigned to one of the above

International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36


13
groups depending on the given splicing pattern. which is known to be an interaction module for specific
Unexpectedly, a large number of miRNAs are found in proline-rich sequences. It remains to be determined if this
introns of protein-coding genes (90 out of 161 miRNAs). domain interacts with the proline rich region of Drosha.
This indicates that previous Bio-informatic searches confined Although the biochemical role of DGCR8/Pasha is currently
to intergenic regions might have missed some miRNA genes. unclear, it is believed to assist Drosha in substrate
The location of some intronic miRNAs is well conserved recognition. There are hundreds of different pri-miRNAs in
among diverse species. For instance, miR-7 is found in the animal cells, but they do not seem to share any common
intron of hnRNPK in both insects and mammals. Another sequence motifs. This question cannot be fully answered yet,
interesting example is the miR-106bw25w93 family that is but mutagenesis analyses indicate that the tertiary structure of
found in the intron 13 of MCM7 in both humans and mice. pri-miRNAs is the primary determinant for substrate
As expected for genes sharing the same promoters, the ‗host‘ specificity 58, 59. Both the double-stranded stem structure
transcript and miRNAs usually have similar expression around the cleavage site and the large terminal loop
profiles. MiRNA promoters have been identified (consisting of >10 nucleotides) are vital. Interestingly, it
experimentally in numerous studies42-48. Bio-informatic seems that the Drosha complex can measure the length of the
searches for miRNA-specific promoter elements upstream of stem, because the cleavage site is located approximately two
miRNA sequences have not been successful. Instead, the helical turns (~22 nucleotides) from the terminal loop. To
characterized miRNA promoters contain general RNA comprehend how the Drosha complex measures the length,
polymerase II (Pol II) transcriptional regulatory elements the biochemical roles of the individual domains of Drosha
previously found in protein coding genes. and DGCR8 need to be delineated.
2.2 Nuclear export by exportin-5
2. Biogenesis of Micro RNA: Following nuclear processing by Drosha, pre-miRNAs are
The current model for miRNA maturation was formulated on exported to the cytoplasm. Once there, they are subjected to
the basis of two simple observations49.First, miRNAs are the second processing step by Dicer (another RNase III
transcribed as long primary transcripts that are first trimmed enzyme) to generate the final ~22-nucleotide product. Owing
into the hairpin intermediates (pre-miRNAs) and to compartmentalization of the two processing events, nuclear
subsequently cleaved into mature miRNAs. Second, the export of pre-miRNAs is a crucial step in miRNA biogenesis
60,61
catalytic activities for the first and the second processing are . Nuclear transport occurs through nuclear pore
compartmentalized into the nucleus and the cytoplasm, complexes, which are large proteinaceous channels
respectively (Figure-4). So the nuclear export of pre-miRNA embedded in the nuclear membrane62. Members of the
is necessary for the cytoplasmic processing to occur. nuclear export receptor family bind cooperatively to a cargo
as well as to the GTP-bound form of the cofactor Ran in the
2.1 Nuclear processing by Drosha nucleus. Following export, hydrolysis of GTP to GDP results
Transcription of miRNA genes yields primary transcripts, in release of the cargo from the export complex. Export of
pri-miRNAs, that are usually several kilo-bases long and that pre-miRNA is mediated by one of the nuclear transport
contain a local hairpin structure. The stem-loop structure is receptors, exportin-563-65. When the cells were depleted of
cleaved by the nuclear RNase III Drosha to release the exportin-5, the pre-miRNA level and the mature miRNA
precursor of miRNA (pre-miRNA)50 (Figure-3a) The level were reduced in the cytoplasm. Notably, pre-miRNA
remnants (the flanking fragments) are thought to be degraded does not accumulate in the nucleus subsequent to the
in the nucleus, although the ~1.7-kb 3′ flanking fragment of depletion of exportin-5. This indicates that pre-miRNA might
miR-23a~27a~24-2 has been cloned multiple times in EST be relatively unstable and also that pre-miRNA might be
studies and is currently annotated as an stabilized through its interaction with exportin-5. Exportin-5
mRNA51.Accordingly, it remains to be seen whether the 5′ was originally known as a minor export factor for tRNAs,
and 3′ fragments that surround the stem-loop have their own because it can transport tRNAs when the primary export
functions. Drosha is a large protein of ~160 kDa, and is factor, exportin-t, is depleted or overloaded66,67.Considering
conserved in animals52-54. It contains two tandem RNase III that the affinity of exportin-5 to pre-miRNA is much higher
domains (RIIIDs) and a double-stranded RNA-binding than to tRNA, and that miRNAs are as abundant as 50,000
domain (dsRBD) that are crucial for catalysis55. The central copies per cell68, pre-miRNAs are probably the main cargo
region of the protein, adjacent to the RIIIDs, is also essential for exportin-5.
for pri-miRNA processing55.Drosha forms a large complex of
~500 kDa in D. melanogaster 56 or ~650 kDa in humans55,57 In 2.3 Cytoplasmic processing by Dicer
this complex, which is known as the Microprocessor Following their export from the nucleus, pre-miRNAs are
complex, Drosha interacts with its cofactor, the DiGeorge subsequently processed into ~22-nucleotide miRNA duplexes
syndrome critical region gene 8 (DGCR8) protein in humans by the cytoplasmic RNase III Dicer 69-73(Figure-3b). Because
(also known as Pasha in D. melanogaster and C. Dicer was originally found to function in generating siRNAs,
elegans).DGCR8/Pasha is a ~120 kDa protein that contains which are similar in size (21–25 nucleotides) to miRNAs, it
two dsRBDs. It also contains a putative WW DOMAIN, was predicted that Dicer also functions in the processing of

International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36


14
Figure 5. Mechanisms of miRNA-mediated repression
Mature miRNAs are incorporated into effector complexes
that are known as ‗miRNP‘(miRNA-containing
ribonucleoprotein complex), ‗mirgonaute‘ or ‗miRISC‘
(miRNA-containing RNA-induced silencing complex).On the
other hand, the effector complex that contains siRNA is
Figure 4. The biogenesis of microRNAs. known as ‗RISC‘, ‗sirgonaute‘ or ‗siRISC‘.The cleavage
~70-nucleotide stem-loop RNAs into mature miRNAs. products (~22-nucleotide miRNA duplexes) do not persist in
Indeed, it was later proven that immuno-precipitated Dicer the cell for long. Usually, one strand of this short-lived
generates ~22-nucleotide miRNAs from in vitro synthesized duplex disappears, whereas the other strand remains as a
~70-nucleotide let-7 stem-loop RNAs. Moreover, when the mature miRNA. Studies on siRNA duplexes indicate that the
human homologue and C. elegans homologue of Dicer were relative Thermo-dynamic stability of the two ends of the
knocked down or knocked out, the ~70-nucleotide pre- duplex determines which strand is to be selected88,89.The
miRNAs accumulated, whereas the ~22-nucleotide mature strand with relatively unstable base pairs at the 5′ end
miRNAs diminished. Dicer is a highly conserved protein that typically remains (for example, G:U pair versus G:C
is found in almost all eukaryotic organisms such as pair)88,89. The same rule is thought to be applicable to
Schizosaccharomyces pombe, plants and animals. Some miRNA. The mechanistic details of strand selection have
organisms contain multiple Dicer homologues, in which been delineated in D. melanogaster. R2D2, which has two
different Dicer isotypes are often assigned to take on distinct dsRBDs, was shown to sense the differences in
roles. For example, D. melanogaster Dicer-1 is required for thermodynamic inequalities. R2D2 forms a stable
pre-miRNA cleavage, whereas Dicer-2 is needed for siRNA heterodimeric complex with Dicer-2 and binds to the more
generation. Dicer homologues are multi-domain proteins of stable end of the siRNA duplex, thereby orienting the
~200 kDa .Apart from two RIIIDs and a dsRBD, Dicer has a complex on the siRNA duplex. It is not known how the
long N-terminal segment that contains a DEAD-BOX RNA unselected strand is removed and degraded. It also remains to
helicase domain, as well as a DUF283 domain and a PAZ be determined if the same machinery acts on the miRNA
domain. The PAZ domain is also found in a group of highly duplex and how conserved this machinery is.
conserved proteins known as Argonaute proteins. Structural
and biochemical studies of the PAZ domain from D. 2.4 Collaborative actions of biogenesis factors
melanogaster AGO1 and AGO2 indicate that the PAZ The multiple steps in miRNA biogenesis seem to be
domain binds to the 3′ protruding end of small RNAs74-77. remarkably well coordinated. Drosha initiates miRNA
The roles of the other domains in Dicer remain unknown. processing by the specific cropping of the stem-loop
Dicer associates with several other proteins including RDE-4 precursor in the nucleus90. Like other RNase-III-type
in C. elegans78, R2D279 and FMR180–83 in D. melanogaster 80, endonucleases, Drosha cuts dsRNA to create a short ~2-
and Argonaute family proteins in various organisms84,85. nucleotide 3′ overhang90,91. The resulting structure (a short
These Dicer-interacting proteins do not seem to be required stem plus a 3′ overhang) seems to be a signature motif for all
for the cleavage reaction itself as purified human Dicer and dsRNAs that are involved in small-RNA pathways. Exportin-
D. melanogaster Dicer-2 can catalyse the cleavage 5 recognizes this signature motif to export pre-miRNA to the
reaction80,86, 87. Instead, Dicer-interacting proteins have cytoplasm. Following export, pre-miRNA is handed over to
various roles in miRNA stability and effector complex another RNase III Dicer, which has a preference for the
formation and action. For example, one of the Argonaute terminus of dsRNAs containing the short 3′ overhang92.
family proteins, human AGO2, was recently shown to Therefore, Drosha confers specificity to the biogenesis
function as the ‗slicer‘ enzyme that cleaves target mRNA. pathway and facilitates the overall production rate by
International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
15
2.5 MicroRNA biogenesis in plants
Homologues of Drosha and DGCR8/Pasha have not been
found in plants, which indicate that the Drosha dependent
stepwise processing model applies only to animal cells.
Neither miRNA nor the Drosha homologue has been found in
yeasts. Plant miRNA precursors are quite diverse in structure,
and the stem-loops are usually longer than animal pri-
miRNAs. Genetic studies in A. thaliana showed that DCL1,
one of the four Dicer like proteins in A. thaliana (also known
as CARPEL FACTORY (CAF)), is important for miRNA
accumulation94–95. DCL1 is a nuclear protein, which
indicates that mature ~22-nucleotide miRNA might be
generated in the nucleus in plants96. Although detection of
miRNA precursors has been difficult, presumably owing to
efficient processing and/or rapid turnover of precursors, at
least one study showed that plant miRNAs, similar to animal
miRNAs, might be processed in a stepwise manner97, in
which DCL1 might be solely responsible for all processing
steps. HASTY (HST) is a plant homologue of exportin-5 on
Figure 6. MicroRNAs can function as tumour suppressors the basis of its amino-acid sequence. An HST mutant showed
and oncogenes pleiotropic phenotypes, which indicates that this protein
might function in miRNA biogenesis too98, 99. A recent
(a) In normal tissues, proper microRNA (miRNA) transcription,
study showed that loss-of-function mutants in HST reduced
processing and binding to complementary sequences on the target
mRNA results in the repression of target-gene expression through a the accumulation of most miRNAs, indicating that HST
block in protein translation or altered mRNA stability (not shown). functions as a nuclear export receptor100. The identity of the
The overall result is normal rates of cellular growth, proliferation, actual cargo for HST-whether it is precursor, miRNA duplex
differentiation and cell death. or single-stranded miRNA is unclear. Additional miRNA
(b) The reduction or deletion of a miRNA that functions as a tumour biogenesis factors include HYL1, a two dsRBD-containing
suppressor leads to tumour formation. A reduction in or elimination nuclear protein of unknown biochemical function101, 102,
of mature miRNA levels can occur because of defects at any stage of and HEN1, a protein with a dsRBD and a methyl-transferase
miRNA biogenesis (indicated by question marks) and ultimately domain103, 104. It was recently shown that HEN1
leads to the inappropriate expression of the miRNA-target
methylates miRNA duplex at the 2′ hydroxyl groups of the 3′-
oncoprotein (purple squares). The overall outcome might involve
increased proliferation, invasiveness or angiogenesis, decreased end nucleotides104.The biochemical role of this 2′-O-methyl
levels of apoptosis, or undifferentiated or de-differentiated tissue, group awaits further investigation.
ultimately leading to tumour formation.
(c) The amplification or over-expression of a miRNA that has an 2.6 Regulation of microRNA biogenesis :
oncogenic role would also result in tumour formation. In this Expression profiling studies indicate that most miRNAs are
situation, increased amounts of a miRNA, which might be produced under the control of developmental and/or tissue specific
at inappropriate times or in the wrong tissues, would eliminate the signaling pathways105-115. miRNA expression might be
expression of a miRNA-target tumour-suppressor gene (pink) and regulated at multiple steps of RNA biogenesis, although it
lead to cancer progression. Increased levels of mature miRNA might
remains to be determined which step is controlled and how
occur because of amplification of the iRNA gene, a constitutively
active promoter, increased efficiency in miRNA processing or this control is achieved. Transcriptional regulation is
increased stability of the miRNA (indicated by question marks). probably the main control mechanism. For example, the
ORF,open reading frame. temporal regulation of let-7RNA in C. elegans is dependent
generating the 3′ protruding ends, which are recognized efficiently on a transcriptional enhancer element, known as the temporal
by the downstream biogenesis factors92. In addition, Drosha pre- regulatory element (TRE)116. This element is situated ~1200
determines mature miRNA sequences by precisely generating one bp upstream of the mature let-7 RNA. Electrophoretic
end of the mature miRNA90, 93. The other end is created by Dicer mobility shift assays using nuclear extract indicated that a
and measures ~22 nucleotides from the pre-existing terminus of the ~22-nucleotide inverted repeat in the TRE might be a binding
pre-miRNA RNA interference (RNAi)-mediated knock-down of
site for a transcription factor that is yet to be identified116.
Drosha, exportin-5 or Dicer results in a significant reduction, but not
the full loss, of mature miRNA .This is probably due to the long Some miRNAs seem to be controlled at the
half-life of mature miRNA. miRNAs seem to be quite stable once posttranscriptional level. For example, C. elegans miR-38 is
they enter into the effector complex. But the possibility that there expressed only in the embryo, whereas the precursor (pre-
could be minor alternative biogenesis pathway(s) cannot be miR-38) is ubiquitously detected, which indicates that the
excluded. For example, a small portion of pri-miRNAs might be maturation of pre-miR-38 might be temporally regulated. It is
exported by mRNA export pathway and become processed in the possible that pre-miR-38 is retained in the nucleus until
cytoplasm.
International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
16
export is triggered by a certain developmental signal. 3. The mechanism of action of microRNAs:
Alternatively, it might be bound to negative regulatory The mechanism of action of microRNAs is considered to be
factor(s) in the cytoplasm, so that they remain inaccessible to by two modes – translational repression and target
Dicer until a certain stage. degradation (Figure-5). The former is common in mammalian
systems while the latter is found predominantly in plants. The
2.7 Computational tools for discovery of microRNA and basic difference in the two mechanisms is thought to be
their targets: primarily governed by the levels of complementarity between
Computational predictions have been the mainstay for microRNAs and their target transcripts. Perfect or near
discovery of microRNAs and their targets. The algorithms for perfect complementarity as is common in plant microRNAs
microRNA prediction range from custom-made programs to and in a small class of eukaryotic microRNAs causes target
search for hairpin loops and energetic stability to advanced cleavage and degradation131analogous to the action of
algorithms employing machine learning approaches117-119. siRNAs which have perfect complementarity to the target
Even though the algorithms for microRNA prediction have regions. Evidence suggests that microRNA bound transcripts
improved over time, accurate de novo prediction of are sequestrated into P bodies132, 133 where they are
microRNA still remains a challenging task. This is especially maintained in a silenced state either by associating with
important in the case of viruses as viral microRNAs do not proteins that prevent translation or possibly by removal of the
share close homology even in the same class. We hope the cap structure 133.
prediction algorithms will improve with a better
understanding of the sequence and structural components of 3.1 Translational inhibition:
precursor hairpins involved in microRNA biogenesis120. The The first mechanistic analyses of miRNA function were
algorithms for microRNA target prediction also have been carried out using C. elegans: it was found that the abundance
significantly improved from first-generation algorithms of miRNA-regulated mRNAs was not substantially hanged,
which rely on sequence complementarity rules, but the abundance of proteins encoded by those mRNAs was
thermodynamic stability and conservation121, 122 by markedly reduced 134,135. Furthermore, the regulated
incorporation of features like target RNA structure123. mRNAs seemed to be present in polysomes134,135 and
2.8 miRNAs are a large family of gene regulators: engaged with ribosomes capable of subsequent elongation in
miRNAs are non-coding, single-stranded RNAs of ~22 vitro135 .These striking observations suggested that miRNA-
nucleotides and constitute a novel class of gene regulators mediated suppression of protein production occurred by a
that are found in both plants and animals. They negatively mechanism that operated after the initiation of protein
regulate their targets in one of two ways depending on the synthesis. Although subsequent studies have shown that
degree of complementarity between the miRNA and the miRNAs can exert their effects by a variety of mechanisms,
target. First, miRNAs that bind with perfect or nearly perfect there are now numerous examples in which miRNA-mediated
complementarity to protein coding mRNA sequences induce reduction of protein production is not accompanied by
the RNA-mediated interference (RNAi) pathway. Briefly, corresponding changes in mRNA abundance136-140. In most
mRNA transcripts are cleaved by ribonucleases in the of these cases, the translational status of regulated mRNAs
miRNA-associated, multi-protein RNAinduced- silencing (i.e.whether they are associated with polysomes) has not been
complex (miRISC), which results in the degradation of target determined. Nevertheless, three recent reports provide
mRNAs. This mechanism of miRNA-mediated gene evidence for repression of protein production after protein
silencing is commonly found in plants 124-126, but miRNA- synthesis has been initiated141-143.In the first study, a
directed mRNA cleavage has also been shown to occur in miRNA-regulated mRNA in HeLa cells was found
mammals127. However, most animal miRNAs are thought to exclusively in polysomes and associated with elongationally
use a second mechanism of gene regulation that does not competent ribosomes141. In the second study, a reporter
involve the cleavage of their mRNA targets. These miRNAs mRNA responsive to an endogenous miRNA was also
exert their regulatory effects by binding to imperfect present in polysomes142. Several lines of evidence showed
complementary sites within the 3′ untranslated regions that these polysomes were actively translating; however,
(3‘UTRs) of their mRNA targets, and they repress target- nascent polypeptides could not be detected by immuno-
gene expression post-transcriptionally, apparently at the level precipitation, suggesting that repression of protein
of translation, through a RISC complex that is similar to, or accumulation resulted from co-translational protein
possibly identical with, the one that is used for the RNAi degradation (i.e. protein degradation concomitant with
pathway128-129. miRNAs that use this mechanism reduce translation). In the third study, a reporter constructs
the protein levels of their target genes, but the mRNA levels containing a 3‘-UTR with designed target sites for a synthetic
of these genes are barely affected. However, recent findings miRNA that has extensive but imperfect complementarity to
indicate that miRNAs that share only partial complementarity those sites was shown to be translationally repressed and
with their targets can also induce mRNA degradation, but it is associated with polysomes143. In this case, evidence
unclear if translational inhibition precedes destabilization of suggested that the deficit in protein production resulted from
the gene targets in these cases130. premature termination of translation (premature ribosome

International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36


17
Table-1: microRNAs and their targets: examples of microRNAs with experimentally validated functions/targets
Micro RNA Target Gene Function Modes of References
Repression
Caenorhabditis elegans
lin-4 lin-14,lin-28 Regulation of developmental transition Translational 173,174,175
between the first two larval stages, L1 repression
and L2
let-7 lin-41, hbl-1 Regulation of developmental transition Translational 176-179
between the last larval stage (L4) and the repression
adult stage
lsy-6 cog-1 Determination of left/right asymmetry of Unknown 180
neuronal development
Drosophila melanogaster
Bantam hid Promotion of cell proliferation and Translational 181
suppression of apoptosis repression
Mus musculus

miR-181 Unknown Promotion of haematopoietic Unknown 183


differentiation towards the B-cell lineage.
miR-196 Hoxb8 Unknown PTGS 184
Arabidopsis thaliana
miR-165, miR- PHB, PHV and Regulation of leaf morphogenesis PTGS 185, 186
166 REV
miR-172 AP2 Regulation of flowering time and floral- Translational 187
organ identity repression
miR-JAW TCP Regulation of leaf development and PTGS 188
transcription embryogenesis
factors
miR-39 SCLfamily Unknown PTGS 189
proteins
Zea mays
miR-166 rid1 Regulation of leaf morphogenesis PTGS 190
AP2, APETALA 2; Hid, head involution defective; Hoxb8, homeobox B8; PHB, PHABULOSA; PHV, PHAVOLUTA; PTGS,
post-transcriptional gene silencing; REV,REVOLUTA; rld1, rolled leaf1; SCL, SCARECROW-LIKE; TCP, teosinte branched
1-cycloidea-PCF.
drop off) 143. By contrast, two other reports have showed that it was impaired143-145. The most
demonstrated miRNA-mediated inhibition at the level of straightforward interpretation of the results obtained so far is
initiation of protein synthesis144, 145. In one study, an that miRNA-mediated inhibition of translation can occur in
endogenous miRNA was shown to reduce polysome an mRNA-specific manner at distinct steps in the process of
association of a reporter construct with appropriate target translation. Nevertheless, the features of individual mRNAs
sites144. In the other study, complete repression of and/or mRNA–miRNA combinations that dictate the
translation required both the 5‘-cap and the 3‘-poly (A)+ tail, mechanism of repression remain to be elucidated.
whereas internal ribosome entry site (IRES)-directed
translation was insensitive to inhibition145. Although the 3.2 mRNA de-adenylation and degradation:
results of these two studies differ in the details [e.g. the role It was originally thought that miRNA-mediated regulation
of the poly (A)+ tail], they both present persuasive evidence was exerted mainly at the level of translation and not at the
that initiation of protein synthesis is impaired by miRNAs at level of mRNA degradation. It is now clear that this idea is
the level of cap recognition. Collectively, the studies of only partially correct. There are numerous examples of
translational inhibition have revealed distinct mechanisms by miRNAs destabilizing their target mRNAs. Indeed, as
which cap-dependent translation can be repressed. detailed in this section, the mechanism by which miRNAs
Interestingly, differences have also been observed in the increase mRNA turnover is perhaps better understood than
ability of miRNAs to suppress translation promoted by the mechanism of translational repression. Early evidence for
miRNA-mediated destabilization came from studies in which
IRESs: two groups found that IRES-directed protein a miRNA was transfected into cells that normally did not
synthesis was unaffected by miRNAs, whereas another group express this miRNA. The presence of such a miRNA was

International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36


18
Table 2: MicroRNAs that are associated with human cancers
miRNA Gene loci Cancer association Function References
miR-15a, Chromosome Frequently deleted or downregulated in TS 209,210
miR-16-1 13q14 B-cell chronic lymphocytic leukemia;
negatively regulates the antiapoptotic
gene BCL2
miR-143, Chromosome Decreased abundance in colorectal TS 211,212
miR-145 5q32–33 cancer; downregulated in breast,
prostate, cervical and lymphoid cancer
cell lines; miR-145 is decreased in
breast cancer.
miR-21 Chromosome Anti-apoptotic factor; upregulated in OG 211,213,214
17q23.2 glioblastomas and breast cancer.
let-7 Multiple loci Negatively regulate the Ras oncogenes; TS 215,216
family direct cell proliferation and
members differentiation; decreased abundance in
lung cancer
miR-142 Chromosome A t(8;17) translocation that places the N/A 217
17q22 MYC oncogene downstream of the
mir-142 hairpin, resulting in an
aggressive
B-cell leukemia that is due to MYC
overexpression
BIC/miR- Chromosome Upregulated in paediatric Burkitt, OG 211,218-221
155 21q21 Hodgkin, primary mediastinal and
diffuse large-B-cell lymphomas;
upregulated in human breast cancer
miR-17– Chromosome Upregulated by MYC; negatively TS/ OG 222,223
19b cluster 13q31–32 modulates the E2F1 oncogene; loss
of heterozygosity of this cluster is
found in hepatocellular carcinoma;
overexpressed in B-cell lymphomas
N/A-not applicable; OG-oncogene; TS- tumour suppressor.

found to reduce the abundance of many mRNAs, most of three reporter mRNAs were analyzed. Although all three
which contained target sites for the miRNA146. In addition, were completely de-adenylated, one remained stable, a
examination of several miRNA targets in C. elegans revealed second was only partially destabilized, and turnover of a third
that these also were reduced in abundance in a miRNA- was markedly increased. Importantly, the level of regulation
dependent manner147. The realization that miRNAs can of these reporter mRNAs did not correlate with their stability
promote mRNA degradation has enabled the use of mRNA in all cases: that is, the stable de-adenylated RNA was still
microarray analysis for the identification of miRNA strongly repressed at the translational level149. Although the
targets146, 148-150. Although this approach has been translational repression observed could have resulted from
fruitful, the proportion of the total number of targets that is the loss of the polyA tail, maintaining the poly(A)+ tail (by
identified by these types of analysis remains to be knockdown of the de-adenylase) did not relieve the
determined. There is no analogous high throughput method to inhibition149. Results from human cells, showing miRNA-
identify targets that are regulated strictly at the translational dependent translational inhibition of a non-polyadenylated
level. It now seems clear that miRNA-mediated mRNA, are also inconsistent with de-adenylation being the
destabilization is a consequence of de-adenylation followed sole cause of repression of protein synthesis152.
by decapping (i.e. removal of the 5‘-cap). Many studies – in Furthermore, repression of protein synthesis is not
zebrafish embryos, human cells and Drosophila melanogaster responsible for deadenylation: two groups have shown that
cells – have shown miRNA-dependent de-adenylation149, mRNAs that cannot be translated, because of either a strong
151-153. In D. melanogaster cells, de-adenylation is carried stem–loop in the 50-UTR or a defective cap, are still subject
out by the de-adenylase complex CCR4–NOT. Interestingly, to deadenylation152, 153. These results from the study of
although de-adenylation seems to be a prerequisite for mRNA several species and many distinct mRNAs suggest that
decapping and degradation, it does not necessarily lead to increased de-adenylation might be a general consequence of
these outcomes. Specifically, in the study in D. melanogaster, miRNA–mRNA interactions. However, no change in

International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36


19
poly(A)+ tail length was observed for an miRNA regulated Secondly, when human miRNAs were discovered, it was
mRNA in C. elegans, although in this case the tail was short noticed that many miRNA genes were located at fragile sites
under all conditions examined. It will be of considerable in the genome or regions that are commonly amplified or
interest to determine whether other miRNA-regulated deleted in human cancer193. Thirdly, malignant tumors and
mRNAs are de-adenylated, particularly those that seem to be tumor cell lines were found to have widespread deregulated
repressed by distinct mechanisms and those for which miRNA expression compared to normal tissues194-
miRNA-mediated repression is reversible154. 196(Table-2). The question remained whether the altered
miRNAs regulate multiple gene targets (Table-1). miRNA expression observed in cancer is a cause or
The current challenge is to accurately identify targets that are consequence of malignant transformation.
regulated by miRNAs. Because miRNAs usually bind to their
targets with incomplete complementarity, which allows short 4.1 MicroRNAs as causal cancer genes at genomic
stretches of mismatched base-pairs and G–U base pairing, the breakpoints :
identification of gene targets with a simple BLAST search is Five years ago, the first direct evidence for an involvement of
impossible. However, current bio-informatic approaches have miRNAs in cancer was reported197. Calin et al. studied a
taken advantage of the fact that miRNAs within families have well-known deletion on chromosome13, which is the most
highest homology at the 5′ end of the mature miRNA. Several frequent chromosomal abnormality in chronic lymphocytic
studies have indicated that the 5′ end of the miRNA is crucial leukemia (CLL). This deletion had long been suspected to
for the stability and proper loading of the miRNA into the contribute to leukemogenesis. However, extensive studies
miRISC complex155, 156, and this end is also important for had failed to identify a causal gene. Calin et al197found that
biological function157–159. Therefore, most bioinformatics two miRNA genes, mir-15 and mir-16, were located within
algorithms use a ‗miRNA seed‘ that encompasses the first 2– this 30-kb deletion. They subsequently analyzed the
8 bases of the mature miRNA sequence to search for expression of miR-15 and miR-16 in blood samples from
complementarity to sequences in the 3′ UTR of all expressed patients with CLL. Both miRNAs were absent or down-
genes. These studies have revealed that a single miRNA regulated in the majority (68%) of cases when compared to
might bind to as many as 200 gene targets and that these normal tissue or lymphocytes. This finding suggested that
targets can be diverse in their function; they include these two miRNAs were causally involved in the
transcription factors, secreted factors, receptors and pathogenesis of chronic lymphocytic leukemia. In 2005, three
transporters160-169.So, miRNAs potentially control the reports provided the first mechanistic insight into how
expression of about one-third of human mRNAs. However, miRNAs might contribute to carcinogenesis. Two
many targets are missed using the ‗miRNA seed‘ approach as independent studies described the relationship between a
there is evidence that other variables besides the first 2–8 miRNA cluster, mir-17-92, and the Myc oncogenic
nucleotides of the miRNA sequence dictate miRNA pathway198, 199. A third report demonstrated an interaction
regulation. For example, the complete mature miRNA between let-7 miRNA and the RAS proto-oncogene200
sequence of let-7 has been evolutionarily conserved from
worms to humans170, demonstrating the functional relevance 4.2 MicroRNAs with oncogenic Potential:
of the 3′ end. Furthermore, mutagenesis studies in one A cluster of six miRNAs, the mir-17-92 cluster, was found to
laboratory have shown that base pairing at both the 5′ and 3′ be located within a region on chromosome 13 that is
ends of let-7 is required for down regulation of a target commonly amplified in human B-cell lymphomas201. He et
gene171. Therefore, the number of known miRNA targets al.202demonstrated that the miRNAs from the mir-17-92
could increase and the challenge will be to determine which cluster were over expressed in lymphoma cell lines carrying
predicted targets are biologically relevant. Sites that are this amplification, and expression levels correlated with gene
complementary to several miRNAs have also been identified copy number of the mir-17-92 locus. Further, the miR-17-92
within the 3′ UTR of a single gene target, indicating that primary transcript was found to be over expressed in tumor
complex patterns of combinatorial regulation by miRNAs samples from lymphoma patients. To test their hypothesis
exist172. Because miRNAs potentially have a broad that mir-17-92 actively contributes to lymphomagenesis, the
influence over several diverse genetic pathways, the deletion authors took advantage of a mouse model of human B-cell
or mis-expression of these small RNAs is likely to be lymphoma. These mice develop lymphomas due to an over
pleotropic and contribute to disease, including cancer. expression of the Myc oncogene. The Myc oncogene encodes
the transcription factor c-Myc that regulates cell proliferation,
4. MicroRNAs and cancer: growth, and apoptosis, and over expression of c-Myc is
Three important observations early in the history of miRNAs common in cancer. He et al. demonstrated that additional
suggested a potential role in human cancer. Firstly, the expression of the mir-17-92 cluster accelerated c-Myc-
earliest miRNAs discovered in the roundworm C. elegans and induced tumorigenesis in mice. The authors therefore
the fruit fly Drosophila were shown to control cell suggested that mir-17-92 was the first potential non-coding
proliferation and apoptosis191, 192. Their deregulation may oncogene, referred to as oncomir-1.The cellular function of
therefore contribute to proliferative diseases such as cancer. miR-17-92 was not identified in these experiments.

International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36


20
Nevertheless, the pathology of the tumors indicated lower response to stress and cancer-initiating events such as DNA
rates of apoptosis as compared to tumors with Myc over damage. He et al.225 found that a miRNA, miR-34, is
expression alone. Three recent studies contributed towards directly activated by the transcription factor p53 after DNA
our understanding of the oncogenic potential of miR-17-92. damage. Expression of miR-34 induces cell cycle arrest and
Two reports demonstrated an anti-apoptotic effect of miR-17- thereby acts together with other effectors of the p53 tumor
92 through various pathways that promote cell proliferation suppressor network to inhibit inappropriate cell proliferation.
and growth203, 204. A third study identified mir-17-92 as a Another group independently demonstrated that miR-34 is
mediator of angiogenesis in tumors induced by the oncogene up-regulated by p53 upon DNA damage and promotes
c-Myc205.O‘Donnell et al.206 independently identified the apoptosis224. Together, these data indicate that altered
same cluster of miRNAs, mir-17-92, to be regulated by the expression of miRNAs is not simply a secondary event that
transcription factor c-Myc. The transcription factor Myc reflects the less differentiated state of cancer cells. In
induces expression of E2F1 growth factor. The mir-17-92 contrast, at least in some cases, miRNA expression is
cluster which is also induced by c-Myc does, in contrast, specifically driven by tumor suppressors and oncogenes.
inhibit E2F1 expression. The authors therefore suggested a
novel regulatory mechanism by which c-Myc fine-tunes gene 4.5 miRNAs and Tumours of the Nervous System :
expression by activating the transcription of target genes and Several studies found that a high proportion of genomic loci
by simultaneously inducing inhibitory miRNAs that reduce containing miRNA genes exhibit DNA copy number
their translation. The example of the mir-17-92 cluster alterations in common cancers301 and miRNA mis-
highlights that a distinction between oncogenic and tumor expression has also been described in tumours of the nervous
suppressor miRNAs is likely to be an oversimplification. The system and 268–272, 276,302-305. miRNAs have been
same miRNAs may have oncogenic or tumor suppressor shown to act either as tumor suppressors or
activity depending on the context and the cell type they are oncogenes and, depending on the mRNA target, may
expressed in (Figure-6). A single miRNA may regulate accelerate the oncogenic process302. A suppressor effect was
various unrelated target genes and thereby control opposing observed in pituitary adenomas, the most common tumors of
activities such as cellular proliferation and apoptosis. The the central nervous system, in which down-regulation of
ultimate function of a miRNA may depend on the tissue type miR-15a and miR-16 correlates with tumor size 273–275.
they are expressed in and what target genes are present. Other miRNAs, such as the miR-155 and miR17-92 cluster,
have an oncogenic effect306, 307. The consequence of an
4.3 MicroRNAs with tumor suppressor potential : upregulation of miR-21 has been characterized in
The let-7 family of miRNAs was the first group of miRNAs glioblastoma tumor cells268, wherein the knockdown of
shown to regulate expression of a proto-oncogene, the RAS miR-21 led to increased apoptotic cell death, suggesting that
protein. RAS proteins are membrane-associated signaling this miRNA may act as an antiapoptotic player268, 276, 308.
proteins that regulate cell growth and differentiation. A In addition, miRNA profiling in glioblastoma cells has shown
miRNA that controls expression of these potentially high levels of miR-221, miR-128, miR-181a, and miR-181b
oncogenic proteins would be predicted to possess tumor and low levels of miR-181c268, 277, 278.miRNA expression
suppressor activity. Mutations in the RAS oncogene are analysis may also be used for medulloblastoma prognosis.
present in approximately 15–30% of all human cancers, and Down-regulation of miR-9 and miR-125a was observed in
over expression of the RAS oncogene is common in lung aggressive brain malignancy, which results in the activation
cancer. Johnson et al207 showed that over expression of RAS of medulloblastoma cell growth and arrest of apoptosis by
protein in lung cancer tissue correlated with reduced activation of the pro-proliferative truncated TrkC
expression of let-7 miRNA. They experimentally confirmed isoform279.Based on these findings, the potential to
that let-7 can inhibit RAS expression in human cancer cell modulate multiple messages at the same time via miRNA
lines. Loss or reduction of let-7 in lung cancer leads to RAS technology would therefore represent an intriguing prospect
over expression, thus, promoting cellular growth and for cancer treatment.
contributing to tumorigenesis. The authors therefore
suggested that let-7 acts as tumor suppressor207. Another 4.6 MicroRNAs with a role in tumor invasion and
group independently reported reduced expression of let-7 in metastasis:
lung cancers and found that this correlated with a poor Transcriptional networks may drive miRNA expression in
prognosis 208. cancers. Recent work from Ma et al.226 suggested a model
by which a pleiotropic transcription factor, Twist, induces
4.4 MicroRNAs in the p53 tumor suppressor network: expression of a specific miRNA, which suppresses its direct
Transcriptional networks are often deregulated in cancer cells target and in turn activates a pro-metastatic gene, leading to
and may lead to altered transcription of miRNA genes. Two tumor cell invasion and metastasis. The expression of miR-
recent studies identified a miRNA, miR-34, to be regulated 10b induced by the transcription factor Twist promoted cell
by the p53 transcription factor224,225. The p53 protein, also migration and invasion in mouse and human breast cancer
called ―the guardian of the genome‖, regulates the cellular cells. Furthermore, the expression level of miR-10b in

International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36


21
Table-3: miRNAs involved in neurological diseases
microRNA Neurological disease Effect Reference number
miR-29a/b-1 Alzheimer‘s disease downregulation 259
miR-128a Alzheimer‘s disease upregulation 260
miR-298 Alzheimer‘s disease downregulation 261
miR-328 Alzheimer‘s disease downregulation 261
miR-146a Alzheimer‘s disease upregulation 262
miR-133b Alzheimer‘s disease downregulation 263
miR-19 Spinocerebellar ataxia type 1 downregulation 264
miR-101 Spinocerebellar ataxia type 1 downregulation 264
miR-130 Spinocerebellar ataxia type 1 downregulation 264
miR-9 Hungtington‘s disease downregulation 265,266
miR-1 Tourette syndrome deregulation 267
miR-206 Tourette syndrome deregulation 267
miR-21 Glioblastoma upregulation 268
miR-124 Glioblastoma downregulation 269
miR-137 Glioblastoma downregulation 269
miR-124a Medulloblastoma downregulation 270
miR-34a Neuroblastoma downregulation 271
miR-184 Neuroblastoma downregulation 272
miR-15a Pituitary adenoma downregulation 273-275
miR-16 Pituitary adenoma downregulation 273-275
miR-221 Glioblastoma upregulation 268,276-278
miR-128 Glioblastoma upregulation 268,276-278
miR-181a Glioblastoma upregulation 268,276-278
miR-181b Glioblastoma upregulation 268,276-278
miR-181c Glioblastoma downregulation 268,276-278
miR-9, miR-125a Medulloblastoma downregulation 279

primary human breast carcinomas correlated with clinical Based on the differential expression of 217 miRNAs, a
progression226. These findings, if confirmed, suggest that correct diagnosis could be established in 12 out of 17 of the
specific miRNAs may have a role beyond the tumor-initiating tumors. In contrast, gene expression profiling based on
event and directly participate in tumor progression and 16,000 messenger RNAs did not accurately classify the
metastasis.
tumors227. This has potential important clinical implications.
4.7 MicroRNA profiling—implications for cancer If miRNAs prove useful for clinical diagnosis, their key
diagnosis: advantage might be their high stability. In contrast to most
Lu et al227 asked the question whether global miRNA messenger RNAs, they are long-lived in vivo228 and very
expression profiles could classify human cancer. Micro-RNA stable in vitro229, which might allow analysis of paraffin
expression profiles clearly differentiated human cancers embedded samples for routine diagnostic applications.
according to their developmental origin. Cancers of epithelial
and hematopoietic origin had distinct miRNA profiles. A 4.8 Regulation of miRNAs in cancer—who regulates the
subgroup of gastrointestinal tumors, which arise from regulators?
endoderm, was distinguished by miRNA expression patterns. In few cases, the underlying cause of miRNA deregulation in
Furthermore, tumors within a single cell lineage such as acute cancer is clear.The over-expression of miR-17-92 correlates
lymphoblastic leukemia were further differentiated according with amplification of its gene locus230. Similarly, decreased
to their underlying genetic abnormality into BCR/ABL- expression of miR-15 and miR-16 is associated with a
positive tumors, T-cell tumors, and those with MLL gene corresponding chromosomal deletion231. Transcriptional or
rearrangement227. Finally, the authors applied the miRNA epigenetic regulation of miRNAs has been recently reported
expression profiles they had established to an independent 232,233.The transcription of a miRNA gene, mir-124a, was
series of 17 poorly differentiated tumors of unknown origin. shown to be inactivated by hyper-methylation of its promoter
International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
22
in various human tumors. This process of epigenetic silencing plaques (i.e., deposits of Aβ-peptide) 280-284. Only 10%–
is a well-known mechanism to inactivate protein-coding 15% of AD cases represent an inheritable disease which
genes in cancer cells and may similarly apply to miRNAs. follows an autosomal dominant Mendelian pattern, while the
The miRNA gene mir-127 is usually expressed in normal majority arises sporadically. Apparently, the disease may be
cells but not in cancer cells. Saito et al.233 demonstrated that caused by a genetic predisposition, as shown by the
miR-127 was highly induced in cultured human cancer cells identification of specific DNA mutations in a large number of
after treatment with demethylating drugs, suggesting that it is families239, 281-284. Despite a variable etiology, a common
subject to epigenetic silencing through promoter hyper- pathogenetic cascade resulting from distinct gene defects
methylation. A novel mechanism of miRNA regulation was and/or unknown environmental factors cannot be ruled out.
suggested by Mayr et al.234 and Lee and Dutta235. They For example, accumulation of the Aβ peptide, the cause of
demonstrated that miRNA function could be regulated which is unknown, is consistently observed. In approximately
through loss of miRNA binding sites in the target gene. Both 30% of sporadic AD patient samples, the expression of
groups independently demonstrated that chromosomal BACE1 protein, a secretase associated with the formation of
translocations in a known oncogene, high mobility group A2 Aβ-peptide, is significantly increased285.In AD, several
(Hmga2), led to loss of the let-7 miRNA binding sites in its miRNAs exhibit abnormal expression levels, suggesting a
messenger RNA. Disrupted repression of Hmga2 by let-7 dysfunctional orchestration of gene expression 259, 260,
promoted oncogenic transformation and growth in 286,287. Interestingly, Boissonneault et al. have found that
mammalian cells. These two studies provide the first miR-298 and miR-328 bind to the 3‘-UTR of BACE1
evidence that disrupting the interaction of a single miRNA mRNA, thereby producing a regulatory effect on enzyme
and its target can produce an abnormal phenotype in expression in cultured neuronal (N2a) cells. Presence of both
mammalian cells. In addition, there is evidence that miRNAs miR-298 and miR-328 in the hippocampus of APPSwe/PS1
are regulated indirectly through control of their processing mice, a well-documented model for AD, and the observation
enzymes. Thomson et al236 showed that a down regulation of that their levels of expression decrease with aging suggest
miRNAs in human cancer was not associated with reduced that altered levels of these miRNAs may deregulate BACE1
levels of the primary miRNA transcripts. The authors and, in turn, lead to increased Aβ formation and disease
therefore suggested regulation of miRNAs during subsequent progression261. Finally, a recent work from Carrettiero et al.
processing steps, e.g., through altered function of the enzyme shows that miR-128a regulates the cochaperone BAG2 and,
Drosha236. in turn, a pathway of degradation for microtubule-associated
tauproteins with a propensity for misfolding. BAG2 would
5. miRNA and Neurodegenerative Diseases: normally direct tau toward an ubiquitin-independent pathway
Neurodegenerative diseases result from dysfunction, and selectively reduce the levels of sarkosyl-insoluble
progressive deterioration, and extensive loss of neurons in the protein288. Thus, the observation that miR-128a is
central and/or peripheral nervous system237,238. In this upregulated in AD260 may highlight a molecular mechanism
regard, Alzheimer‘s disease (AD)239-242,Parkinson‘s that underlies tau inclusions in neurodegeneration. Taken
disease (PD)243-245, prion diseases246, amyotrophic lateral together, these findings suggest a mechanistic involvement of
sclerosis (ALS)247-248, and hereditary spastic paraplegia249 miRNAs in both the amyloid and tau hypotheses for AD
may have a genetic or sporadic etiology. Instead, pathogenesis.
Huntington‘s disease (HD) 250, 251 and metabolic disorders
with neurological involvement, such as the GM2- 5.2 Parkinson’s Disease (PD) :
gangliosidoses252-257, can only be genetically transmitted. PD is the second most common neurodegenerative disorder,
There is now compelling evidence that disregulation of characterized by resting tremor, muscular rigidity,
miRNA networks is implicated in the development and onset bradykinesia, and impaired balance and coordination243-
of human neurodegenerative diseases258 (Table-3). This, in 245,289-291. Other symptoms include dysautonomia,
turn, may provide the opportunity to elucidate underlying dystonic cramps, and dementia. Typical pathological features
disease mechanisms and open up novel strategies for are loss of dopaminergic neurons in the substantia nigra (SN)
therapeutic applications. and presence of Lewy bodies, which consist of intracellular
inclusions affecting surviving neurons in various areas of the
5.1 Alzheimer’s Disease (AD) : brain289-291. Several gene loci have been implicated in
AD is the most common form of dementia. While several autosomal, dominant forms of PD. These include PARK1 and
hypotheses have been proposed to explain the disease‘s PARK4 (due to a mutation or a triplication of the α-synuclein
etiology, the causes of AD and means of stopping its gene [SNCA] on 4q21 and 4p15, resp.), PARK3 on 2p13,
progression are still elusive matters 239-242,280-284. PARK5 (due to a mutation in the UCHL1 gene) on 4p14,
Features of the disease encompass neuronal loss, PARK8 (due to a mutation in the LRRK2 gene) on 12q12,
intraneuronal neurofibrillary tangles (i.e., aggregates of the PARK10 on 1p, PARK11 on 2q, and PARK13 (due to a
microtubule-associated protein tau following hyper- mutation in the HTRA2 gene) on 2p12289-291,292,293. The
phosphorylation), and extracellular deposits of amyloid implication of miRNAs in PD is intriguing. In murine
International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
23
models, the competence of embryonic stem cells to glutamine segment250, 251, 300.Disruption of miRNA
differentiate into midbrain dopamine neurons in vitro was homeostasis, most likely in connection with an aberrant
shown to be disrupted by Dicer deletion and subsequent functionality of the transcriptional repressor REST, was
suppression of miRNA biogenesis, suggesting a physiological shown to play a dynamic role in HD. In fact, levels of several
role for miRNAs in cell differentiation and/or survival. These miRNAs with upstream RE1 sites are decreased in HD
results were confirmed in vivo, using mice conditional for patient cortices relative to healthy controls. Interestingly, one
Dicer, which exhibited impaired locomotor activity that of these, the bifunctional, brain enriched miR-9/miR-9 targets
recapitulated motility problems observed in PD two components of the REST complex: miR-9 targets REST
patients.Through a subtractive approach, performed by and miR-9 targets CoREST265, 266. As a consequence of a
comparing miRNA expression profiles in normal human markedly altered miRNA expression, target mRNAs are
adult versus PD patients midbrains, it was shown that miR- subject to dysregulated levels which, in turn, affect the
133b is specifically missing in PD and that, based on both physiological status of forebrain neurons265, 266
over-expression and inhibitory tests in vitro, is likely
implicated in the maturation and function of dopaminergic 6. MicroRNAs as an antiviral defense mechanism:
neurons294, 263. A markedly reduced expression of miR- Viruses are obligate intracellular parasites and use the cellular
133b was found in Aphakia mice294, a dopaminergic neuron machinery for their survival and replication. The success of
deficiency model, which lack Pitx3295, a homeobox the virus essentially depends on its ability to effectively and
transcription factor required for neuron survival and normal efficiently use the host machinery to propagate itself. This
motor activity susceptible to polymorphisms associated with dependence on the host also makes it susceptible to the host
sporadic PD296. Together, these observations suggest a gene-regulatory mechanisms. Though the gene regulatory
relationship between miR-133b and Pitx3, which operate mechanisms involving both host and viral proteins have been
through a negative feedback loop, wherein Pitx3 promotes extensively studied, data on small RNA mediated gene
the expression of miR-133b that, in turn, downregulates regulation in viral infections is just emerging. Interestingly, it
Pitx3263. While these results point to a functional role of the seems that the cellular microRNAs, in addition to their
miR-133b/Pitx3 system in ensuring correct dopaminergic normal regulatory roles in cellular gene expression also
function, miR-133b knock-out mice, which are currently double up as fortuitous agents that target foreign nucleic
unavailable, would establish the extent of miR-133b impact acids, as in the case of viruses. The inventory of microRNAs
on PD etiology. On the other hand, another study showed that encoded differs between cell types, and thus may contribute
deletion of Dicer in dopaminoceptive neurons of the murine to the tissue tropism of viruses
striatum led to aberrant anatomical features (smaller brain,
reduced neuron size, astrogliosis) and motor impairments 6.1 Primate Foamy Virus:
(clasping and ataxia) but, surprisingly, not Lecellier et al309, for the first time demonstrated that a
neurodegeneration297. As dysfunction, but not necessarily mammalian microRNA, mir-32 restricts the accumulation of
loss, of dopaminoceptive neurons was previously implicated the retrovirus primate foamy virus type 1 (PFV-1) in human
in PD298, these observations, taken together, suggest that the cells. PFV is a retro-transcribing virus similar to the Human
link between Dicer, miRNAs, and neurodegeneration is Immunodeficiency Virus (HIV), but codes for two additional
restricted to dopaminergic neurons, thereby pointing to proteins, Tas and Bet. Insights into the possible role of
distinct functional roles in dopaminoceptive cells. Finally, microRNAs came from the observation that cell lines which
Wang et al. found that in PD brains and in vitro cell models express a protein, which interferes with the RNA mediated
disruption of the binding site for miRNA- 433 led to silencing machinery, showed higher accumulation of PFV-1.
increased translation of fibroblast growth factor- 20 (FGF20). Disruption of the target site in a mutant of PFV allowed it to
Notably, an FGF20 polymorphism at 8p21.3–22 was accumulate much faster than the wild type, in infected cells.
previously identified as a PD risk factor correlated with The group also demonstrated that Tas could act as a non-
increased α-synuclein expression, and consequently PD specific suppressor of RNA interference and could
onset299. demonstrate that mir-32 related translational block was
indeed higher in Tas(-)cells where Tas was not expressed.
5.3 Huntington’s Disease (HD): This report not only throws light into the role of microRNAs
HD is a fatal, neurodegenerative disorder characterized by in antiviral defense, but also into how viruses have evolved to
involuntary ballistic movements, depression, and offset the effects of RNA interference by encoding
dementia250, 251,300. Hallmarks of HD are progressive suppressors of interference.
chorea, rigidity, and frequent accurrence of seizures,
emotional problems, loss of cognition, as well as atrophy of 6.2 Human Immunodeficiency Virus:
the caudate nucleus. The causal factor of HD is a gene Five human encoded microRNAs can potentially target the
mutation consisting of abnormally extended repeats of the entire repertoire of accessory genes in HIV, including nef.
CAG sequence within the HTT gene, which translates into a The targets were found to be highly conserved in all of the
huntingtin protein containing an excessively increased viral clade sequences with the exception of clade O. The fact
International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
24
that defective nef is well known to be associated with a long associated with virulence. The polymerase activity was
term non-progressor state led us to speculate that the levels of directly correlated with the high virulence of the murine
the cellular microRNAs would be a decisive factor in strain in their cognate host315. Another interesting feature is
determining the progression of the disease. The targets have that these microRNAs were found to be absent in the chicken
been experimentally validated (unpublished results) by genome, although a large number of human microRNAs (160
cloning the target site in the 3'UTR of Green Fluorescent of 336 human microRNAs) have homologs in the chicken
Protein (GFP) reporter gene. Analysis of previously reported genome implicating them in the difference in infectivity and
microarray data310 of these microRNAs in T Cells, lethality of the virus in chicken and human
demonstrated that the microRNA levels are indeed variable
among individuals. Although it is believed that HIV encodes 7. MicroRNAs—novel therapeutic targets?
for suppressors of RNAi311 recent microarray data on Regulatory RNAs may also have therapeutic applications by
microRNA gene expression levels in HIV infected human which disease-causing miRNAs could be antagonized or
cells312 show that above five human encoded microRNAs functional miRNAs restored. The most intuitive choice of
are down regulated. molecules to correct altered miRNA–mRNA interactions are
RNA oligonucleotides. These oligonucleotides need to be
6.3 Mammalian microRNAs as positive regulators chemically modified to allow for stability in serum and
Hepatitis C virus cellular uptake. Modified antisense oligonucleotides are
Jopling et al313 reported an interesting case wherein the already being developed to utilize the intrinsic RNAi pathway
tissue specificity of microRNA expression was exploited by a for delivery of gene therapy. If the delivery problem can be
virus to establish tissue selectivity. A liver specific micro- overcome, then miRNA therapies may also be possible. Two
RNA mir-122 was shown to cause accumulation of viral studies have successfully applied 2′-O-Methylmodified
RNA by binding to the 5' non-coding region of the viral antisense RNAs to inhibit miRNA function in cultured
genome. The authors have also verified the findings by cells316, 317. Recent work by Krutzfeldt et al.318
mutational analysis as well as sequestration of the micro- demonstrated that modified cholesterol-conjugated antisense
RNA. It is possible that this novel mechanism of microRNAs RNAs designated ―antagomirs‖ could effectively inhibit
targeting the 5'UTR of the transcript may be mediated miRNA function in vivo in the adult mouse. The authors
through a translation controlling switch at the 5'end of the applied three daily intravenous injections of antagomirs and
transcript through changes in RNA secondary structure. This, achieved effective inhibition of four miRNAs over a period
by and large, remains the only report of a microRNA of weeks in most tissues except brain. A novel approach was
targeting the 5'UTR in a mammalian system and causing recently reported by Ebert et al 319. They developed miRNA
RNA accumulation. inhibitors that can be transiently expressed in cultured
mammalian cells.These competitive inhibitors termed
6.4 Influenza virus: ―miRNA sponges‖ derepressed miRNA targets at least as
Through computational methods incorporating both strongly as chemically modified antisense
consensus prediction and target accessibility, one group oligonucleotides319. A different approach was taken by
found that human encoded microRNAs could target critical Tsuda et al.320. The authors designed synthetic miRNAs to
genes involved in the pathogenesis and tropism of Influenza target overexpressed tumor proteins, such as HER-2 protein.
virus A/H5N1 (unpublished results). Two human encoded A synthetic miRNA targeting HER-2 messenger RNA
microRNAs mir-507 and mir-136 had potential binding sites successfully inhibited HER-2 protein expression in ovarian
in Polymerase B2 (PB2) and Hemagglutinin (HA) genes cancer cells320. Together, these studies hold some promise of
respectively. The target regions in the respective genes were miRNAs as future therapeutic targets. One limitation of
not only found to be conserved across different viral strains, antisense RNA therapies is the restricted number of cells that
but were also found to fall in highly accessible regions of the can be targeted. Any approach to knockdown a particular
predicted target RNA structure. Moreover, analysis of miRNA with antisense oligonucleotides will only result in
previously reported314 microarray data on microRNA gene partial knockdown. This may represent a limitation for cancer
expression in different tissues has shown that mir-136 is therapies. It remains to be seen whether indirectly mediated
expressed in lung. Both the genes PB2 and HA are known to bystander effects on cancer cells that have not been directly
be critical for the pathogenicity of the virus. While HA is the targeted may partly overcome this limitation. In contrast, a
surface glycoprotein involved in direct binding of the virus to partial effect on function may be of therapeutic value in
the cell surface, HA in the H5N1 subtype carries a polybasic neurodegenerative diseases, such as Parkinson‘s or
site, cleavage at which, by cellular proteases is an essential Alzheimer‘s disease. A partial restoration of dopamine
step in establishing infection. PB2 is one of the three production by antisense therapy might result in a significant
components of the Ribonucleoprotein which is responsible clinical improvement in Parkinson patients. Similarly, a
for RNA replication and transcription. Recent evidence, from partial reduction of the disease-causing proteins in
recombinant viruses generated by combinations of murine Alzheimer‘s disease may lead to a clinical improvement and
and avian viruses identified PB2 as one of the two genes might be achievable by RNA based or miRNA gene therapy.
International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
25
8. MicroRNAs as biomarkers and therapeutics : complex regulatory networks, thereby mediating many facets
Recently microRNA expression profiles have been analyzed of eukaryotic cell function. As Bartel and Chen proposed, the
for viral infections like HIV321. MicroRNA expression has unique combination of miRNAs expressed in each cell type
been shown to be specific to various stages of infection in might affect or ‗dampen‘ the use of thousands of target
Herpesviruses322 and have been proposed to be associated mRNAs. Supporting this hypothesis, the experimental
with latency in HIV infection323, 324, promising an early introduction of muscle- or brain-specific miRNA into HeLa
biomarker for cancers caused by oncogenic viruses. cells shifted the overall mRNA profile to that of muscle or
MicroRNA profiles have also been explored in a number of brain, respectively. Tantalizing correlations between miRNA
patho-physiological conditions325.Recent reports suggest expression and human diseases have been demonstrated.
that microRNA profiles can be used not only to classify Certain miRNAs are modulated in tumors and might be
different classes of cancers326-328, but could also be used as directly involved in the pathology. Understanding the
biomarkers for diagnosis and prognosis of disease states328. miRNA-guided network will provide a new window for
MicroRNAs and anti-microRNA oligonucleotides (AMOs) diagnostics and therapy of many human diseases.
have been proposed as novel therapeutics329,330. Recent
advances in nucleotide chemistry like Locked Nucleic Acids References
(LNA)331, and other backbone modifications have made it
1 Ambros, V. et al. A uniform system for microRNA
possible to design small RNA oligonucleotides which are
annotation.RNA 9, 277–279 (2003).
highly stable in biological systems circumventing one major
2. Kim, V. N. Small RNAs: classification, biogenesis, and
hurdle in using microRNAs as future therapeutics.
function. Mol. Cells 19, 1–15 (2005)
Oligonucleotide modifications have already made their way
3. Xie, Z. et al. Genetic and functional diversification of
to the microRNA experimental biologists‘ workbench332.
small RNA pathways in plants. PLoS Biol. 2, E104 (2004).
The success of siRNA based strategies in targeting specific
4. Vazquez, F. et al. Endogenous trans-acting siRNAs
genes could be extended to microRNAs also. This would
regulate the accumulation of Arabidopsis mRNAs. Mol. Cell
include delivery strategies. This would be even more
16, 69–79 (2004).
important as siRNA based therapeutics for viral pathogens in
5. Peragine, A., Yoshikawa, M., Wu, G., Albrecht, H. L. &
different stages of clinical trials and are showing promising
Poethig, R. S. SGS3 and SGS2/SDE1/RDR6 are required for
results.
juvenile development and the production of trans-acting
siRNAs in Arabidopsis. Genes Dev. 18, 2368–2379 (2004).
9. Artificial microRNAs (amiRNAs) and microRNA
6. Hamilton, A., Voinnet, O., Chappell, L. & Baulcombe, D.
Engineering :
Two classes of short interfering RNA in RNA silencing.
MicroRNAs are promising candidates for developing novel
EMBO J.21, 4671–4679 (2002).
bio-therapeutics against viruses, as it requires only partial
7. Aravin, A. A. et al. The small RNA profile during
complementarity unlike siRNAs and thus can tackle the high
Drosophila melanogaster development. Dev. Cell 5, 337–350
rate of mutations in viruses better than siRNAs. Initial
(2003).
experiments creating microRNAs based on siRNA design
8. Mochizuki, K., Fine, N. A., Fujisawa, T. & Gorovsky, M.
rules have shown promising results333.One group has
A. Analysis of a piwi-related gene implicates small RNAs in
recently developed an algorithm for design of highly specific
genome rearrangement in tetrahymena. Cell 110, 689–699
microRNAs on against sequences to be targeted (unpublished
(2002).
results). This would allow design of microRNAs against
9. Liu, Y., Mochizuki, K. & Gorovsky, M. A. Histone H3
highly conserved sequences in viral genome. Artificial
lysine 9 methylation is required for DNA elimination in
microRNAs also offer the advantage that they can be
developing macronuclei in Tetrahymena. Proc. Natl Acad.
optimized to create less off-target events in the host thus
Sci. USA 101, 1679–1684 (2004).
substantially reducing untoward side effects. MicroRNAs or
10. Taverna, S. D., Coyne, R. S. & Allis, C. D. Methylation
microRNA target sequences could also be engineered into
of histone h3 at lysine 9 targets programmed DNA
transforming viruses and could enable tissue specific and
elimination in tetrahymena. Cell 110, 701–711 (2002).
environment sensitive expression of genes.
11. Ambros, V., Lee, R. C., Lavanway, A., Williams, P. T. &
Jewell, D. MicroRNAs and other tiny endogenous RNAs in
Concluding Remarks:
C. elegans. Curr. Biol. 13, 807–818 (2003).
To date 326 human miRNA genes have been discovered, and
12. Kuwabara, T., Hsieh, J., Nakashima, K., Taira, K. &
there are certainly more to come. A single miRNA can bind
Gage, F. H. A small modulatory dsRNA specifies the fate of
to and regulate many different mRNA targets and,
adult neural stem cells. Cell 116, 779–793 (2004).
conversely, several different miRNAs can bind to and
13. Bartel, D.P. (2004) MicroRNAs: genomics, biogenesis,
cooperatively control a single mRNA target. Two recent
mechanism, and function. Cell 116, 281–297
independent studies predicted that 20–30% of human genes
14. Griffiths-Jones, S. (2004) The microRNA registry.
could be controlled by miRNAs. Thus, hundreds of RNAs
Nucleic Acids Res.32, D109–D111
and thousands of the targets appear to compose remarkably
International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
26
15. Lim, L.P. et al. (2003) The microRNAs of Caenorhabditis 1, controls temporal patterning and is a probable microRNA
elegans.Genes Dev. 2, 2 target. Dev. Cell 4, 639–650 (2003).
16. Chalfie, M., Horvitz, H. R. & Sulston, J. E. Mutations 31. Abrahante, J. E. et al. The Caenorhabditis elegans
that lead to reiterations in the cell lineages of C. elegans. Cell hunchback-like gene lin-57/hbl-1 controls developmental
24, 59–69 (1981). time and is regulated by microRNAs. Dev. Cell 4, 625–637
17. Ambros, V. A hierarchy of regulatory genes controls a (2003)
larvato- adult developmental switch in C. elegans. Cell 57, 32. Slack, F. J. et al. The lin-41 RBCC gene acts in the C.
49–57 (1989) elegans heterochronic pathway between the let-7 regulatory
18. Chalfie, M., Horvitz, H. R. & Sulston, J. E. Mutations RNA and the LIN-29 transcription factor. Mol. Cell 5, 659–
that lead to reiterations in the cell lineages of C. elegans. Cell 669 (2000)
24, 59–69 (1981). 33. Vella, M. C., Choi, E. Y., Lin, S. Y., Reinert, K. & Slack,
19. Ambros, V. & Horvitz, H. R. Heterochronic mutants of F. J.The C. elegans microRNA let-7 binds to imperfect let-7
the nematode Caenorhabditis elegans. Science 226, 409–416 complementary sites from the lin-41 3′ UTR. Genes Dev.
(1984) 18,132–137 (2004).
20. Ambros, V. A hierarchy of regulatory genes controls a 34. Lagos-Quintana, M. et al. Identification of tissue-specific
larvato-adult developmental switch in C. elegans. Cell 57, microRNAs from mouse. Curr. Biol. 12, 735–739 (2002).
49–57. (1989) 35. Sempere, L. F. et al. Expression profiling of mammalian
21. Lee, R. C., Feinbaum, R. L. & Ambros, V. The C. elegans microRNAs uncovers a subset of brain-expressed
heterochronic gene lin-4 encodes small RNAs with antisense microRNAs with possible roles in murine and human
complementarity to lin-14. Cell 75, 843–854 (1993). neuronal differentiation. Genome Biol. 5, R13 (2004).
Described the identification of the first microRNA, lin-4, and 36. Pasquinelli, A. E. et al. Conservation of the sequence and
reported the sequence complementarity between lin-4 and the temporal expression of let-7 heterochronic regulatory RNA.
3′ UTR of the lin-14 mRNA. Nature 408, 86–89 (2000).
22. Wightman, B., Burglin, T. R., Gatto, J., Arasu, P. & 37. Lagos-Quintana, M. et al. (2001) Identification of novel
Ruvkun, G. Negative regulatory sequences in the lin-14 3‘- genes coding for small expressed RNAs. Science 294, 853–
untranslated region are necessary to generate a temporal 858.
switch during Caenorhabditis elegans development. Genes 38. Mourelatos, Z. et al. (2002) miRNPs: a novel class of
Dev. 5, 1813–1824 (1991). ribonucleoproteins containing numerous microRNAs. Genes
23. Ruvkun, G. & Giusto, J. The Caenorhabditis elegans Dev. 16, 720–728
heterochronic gene lin-14 encodes a nuclear protein that 39. Lau, N.C. et al. (2001) An abundant class of tiny RNAs
forms a temporal developmental switch. Nature 338, 313– with probable regulatory roles in Caenorhabditis elegans.
319 (1989). Science 294, 858– 862.
24. Olsen, P. H. & Ambros, V. The lin-4 regulatory RNA 40. Lee, Y. et al. (2002) MicroRNA maturation: stepwise
controls developmental timing in Caenorhabditis elegans by processing and subcellular localization. EMBO J. 21, 4663–
blocking LIN-14 protein synthesis after the initiation of 4670.
translation. Dev. Biol. 216, 671–680 (1999). 41. Rodriguez, A. et al. (2004) Identification of mammalian
25. Ha, I., Wightman, B. & Ruvkun, G. A bulged lin-4/lin-14 microRNA host genes and transcription units. Genome Res.
RNA duplex is sufficient for Caenorhabditis elegans lin-14 14, 1902–1910.
temporal gradient formation. Genes Dev. 10, 3041–3050 42. Tam, W. (2001) Identification and characterization of
1996. human BIC, a gene on chromosome 21 that encodes a
26. Wightman, B., Ha, I. & Ruvkun, G. Posttranscriptional noncoding RNA. Gene 274, 157–167.
regulation of the heterochronic gene lin-14 by lin-4 mediates 43. Aukerman, M.J. and Sakai, H. (2003) Regulation of
temporal pattern formation in C. elegans. Cell 75, 855–862 flowering time and floral organ identity by a microRNA and
(1993) its APETALA2- like target genes. Plant Cell 15, 2730–2741.
27. Moss, E. G., Lee, R. C. & Ambros, V. The cold shock 44. Johnson, S.M. et al. (2003) The time of appearance of the
domain protein LIN-28 controls developmental timing in C. C. elegans let-7 microRNA is transcriptionally controlled
elegans and is regulated by the lin-4 RNA. Cell 88, 637–646 utilizing a Temporal regulatory element in its promoter. Dev.
(1997). Biol. 259, 364–379
28. Pasquinelli, A. E. & Ruvkun, G. Control of 45. Bracht, J. et al. (2004) Trans-splicing and polyadenylation
developmental timing by micrornas and their targets. Annu. of let-7 microRNA primary transcripts. RNA 10, 1586–1594.
Rev. Cell Dev.Biol. 18, 495–513 (2002). 46. Lee, Y. et al. (2004) MicroRNA genes are transcribed by
29. Reinhart, B.J. et al. The 21-nucleotide let-7 RNA RNApolymerase II. EMBO J. 23, 4051–4060.
regulates developmental timing in Caenorhabditis elegans. 47. Cai, X. et al. (2004) Human microRNAs are processed
Nature403, 901–906 (2000).zzzzzzzzzzzzzzzzzzzzzzzzzzz from capped, polyadenylated transcripts that can also
30. Lin, S. Y. et al. The C. elegans hunchback homolog, hbl- function as mRNAs. RNA 10, 1957–1966.

International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36


27
48. Xie, Z. et al. (2005) Expression of Arabidopsis MIRNA elongation factor 1A and tRNA. EMBO J. 21, 6216–6224
genes. PlantPhysiol. 138, 2145–2152. (2002).
49. Lee, Y., Jeon, K. Lee, J. T., Kim, S. & Kim, V. N. 68. Lim, L. P. et al. The microRNAs of Caenorhabditis
MicroRNA maturation: stepwise processing and subcellular elegans. Genes Dev. 2, 991–1008 (2003).
localization. EMBO J. 21, 4663–4670 (2002). 69. Bernstein, E., Caudy, A. A., Hammond, S. M. & Hannon,
50. Lee, Y. et al. The nuclear RNase III Drosha initiates G. J.Role for a bidentate ribonuclease in the initiation step of
microRNA processing. Nature 425, 415–419 (2003). RNA interference. Nature 409, 363–366 (2001).
51. Lee, Y. et al. MicroRNA genes are transcribed by RNA 70. Grishok, A. et al. Genes and mechanisms related to RNA
polymerase II. EMBO J. 23, 4051–4060 (2004). interference regulate expression of the small temporal RNAs
52. Filippov, V., Solovyev, V., Filippova, M. & Gill, S. S. A that control C. elegans developmental timing. Cell 106, 23–
novel type of RNase III family proteins in eukaryotes. Gene 34 (2001).
245, 213–221 (2000). 71.iHutvagner, G. et al. A cellular function for the
53.iFortin, K. R., Nicholson, R. H. & Nicholson, A. W. RNAinterference enzyme Dicer in the maturation of the let-7
Mouse ribonuclease III. cDNA structure, expression analysis, small temporal RNA. Science 293, 834–838 (2001).
and chromosomal location. BMC Genomics 3, 26 (2002). 72. Ketting, R. F. et al. Dicer functions in RNA interference
54. Wu, H., Xu, H., Miraglia, L. J. & Crooke, S. T. Human and in synthesis of small RNA involved in developmental
RNase III is a 160-kDa protein involved in preribosomal timing in C. elegans. Genes Dev. 15, 2654–2659 (2001).
RNA processing. J. Biol. Chem. 275, 36957–36965 (2000). 73. Knight, S. W. & Bass, B. L. A role for the RNase III
55. Han, J. et al. The Drosha–DGCR8 complex in primary enzyme DCR-1 in RNA interference and germ line
microRNA processing. Genes Dev. 18, 3016–3027 (2004). development in Caenorhabditis elegans. Science 293, 2269–
56. Denli, A. M., Tops, B. B., Plasterk, R. H., Ketting, R. F. 2271 (2001).
& Hannon, G. J. Processing of primary microRNAs by the 74. Ma, J. B., Ye, K. & Patel, D. J. Structural basis for
Microprocessor complex. Nature 432, 231–235 (2004). overhangspecific small interfering RNA recognition by the
57.iGregory, R. I. et al. The Microprocessor complex PAZ domain. Nature 429, 318–322 (2004).
mediates the genesis of microRNAs. Nature 432, 235–240 75. Lingel, A., Simon, B., Izaurralde, E. & Sattler, M.
(2004) Nucleic acid 3‘-end recognition by the Argonaute2 PAZ
58. Zeng, Y. & Cullen, B. R. Sequence requirements for domain. Nature Struct. Mol. Biol. 11, 576–577 (2004).
micro RNA processing and function in human cells. RNA 9, 76. Song, J. J. et al. The crystal structure of the Argonaute2
112–123 (2003). PAZ domain reveals an RNA binding motif in RNAi effector
59. Zeng, Y., Yi, R. & Cullen, B. R. Recognition and complexes. Nature Struct. Biol. 10, 1026–1032 (2003).
cleavage of primary microRNA precursors by the nuclear 77. Yan, K. S. et al. Structure and conserved RNA binding of
processing enzyme Drosha. EMBO J. 24, 138–148 (2005). the PAZ domain. Nature 426, 468–474 (2003).
60. Kim, V. N. MicroRNA precursors in motion: exportin-5 78. Tabara, H., Yigit, E., Siomi, H. & Mello, C. C. The
mediates their nuclear export. Trends Cell Biol. 14, 156–159 dsRNA binding protein RDE-4 interacts with RDE-1, DCR-
(2004). 1, and a DExH-box helicase to direct RNAi in C. elegans.
61. Murchison, E. P. & Hannon, G. J. miRNAs on the move: Cell 109, 861–871 (2002).
miRNA biogenesis and the RNAi machinery. Curr. Opin. 79. Liu, Q. et al. R2D2, a bridge between the initiation and
Cell Biol. 16, 223–229 (2004). effector steps of the Drosophila RNAi pathway. Science 301,
62. Nakielny, S. & Dreyfuss, G. Transport of proteins and 1921–1925 (2003).
RNAs in and out of the nucleus. Cell 99, 677–690 (1999). 80. Liu, Q. et al. R2D2, a bridge between the initiation and
63. Lund, E., Guttinger, S., Calado, A., Dahlberg, J. E. & effector steps of the Drosophila RNAi pathway. Science 301,
Kutay, U. Nuclear export of microRNA precursors. Science 1921–1925 (2003).
303, 95–98 (2004). 81. Ishizuka, A., Siomi, M. C. & Siomi, H. A Drosophila
64. Yi, R., Qin, Y., Macara, I. G. & Cullen, B. R. Exportin-5 fragile X protein interacts with components of RNAi and
mediates the nuclear export of pre-microRNAs and short ribosomal proteins. Genes Dev. 16, 2497–2508 (2002).
hairpin RNAs. Genes Dev. 17, 3011–3016 (2003). 82. Caudy, A. A., Myers, M., Hannon, G. J. & Hammond, S.
65. Bohnsack, M. T., Czaplinski, K. & Gorlich, D. Exportin M.Fragile X-related protein and VIG associate with the RNA
5 is a RanGTP-dependent dsRNA-binding protein that interference machinery. Genes Dev. 16, 2491–2496 (2002).
mediates nuclear export of pre-miRNAs. RNA 10, 185–191, 83. Jin, P. et al. Biochemical and genetic interaction between
(2004). the fragile X mental retardation protein and the microRNA
66. Bohnsack, M. T. et al. Exp5 exports eEF1A via tRNA pathway. Nature Neurosci. 7, 113–117 (2004).
from nuclei and synergizes with other transport pathways to 84.iHammond, S. M., Boettcher, S., Caudy, A. A.,
confine translation to the cytoplasm. EMBO J. 21,6205–6215 Kobayashi, R.& Hannon, G. J. Argonaute2, a link between
(2002). genetic and biochemical analyses of RNAi. Science 293,
67. Calado, A., Treichel, N., Muller, E. C., Otto, A. & Kutay, 1146–1150 (2001).
U. Exportin-5-mediated nuclear export of eukaryotic
International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
28
85. Carmell, M. A., Xuan, Z., Zhang, M. Q. & Hannon, G. J. plays a role in microRNA-mediated gene regulation. Proc.
The Argonaute family: tentacles that reach into RNAi, Natl Acad. Sci. USA 101, 1093–1098 (2004).
developmental control, stem cell maintenance, and 103. Boutet, S. et al. Arabidopsis HEN1. A genetic link
tumorigenesis. Genes Dev. 16, 2733–2742 (2002). between endogenous miRNA controlling development and
86. Zhang, H., Kolb, F. A., Brondani, V., Billy, E. & siRNA controlling transgene silencing and virus resistance.
Filipowicz, W.Human Dicer preferentially cleaves dsRNAs at Curr.Biol. 13, 843–848 (2003).
their termini without a requirement for ATP. EMBO J. 21, 104. Yu, B. et al. Methylation as a crucial step in plant
5875–5885 (2002). microRNA biogenesis. Science 307, 932–935 (2005).
87. Zhang, H., Kolb, F. A., Jaskiewicz, L., Westhof, E. & 105. Lagos-Quintana, M. et al. Identification of tissue-
Filipowicz, W. Single processing center models for human specific microRNAs from mouse. Curr. Biol. 12, 735–739
Dicer and bacterial RNase III. Cell 118, 57–68 (2004). (2002).
88. Schwarz, D. S. et al. Asymmetry in the assembly of the 106. Aravin, A. A. et al. The small RNA profile during
RNAi enzyme complex. Cell 115, 199–208 (2003). Drosophila melanogaster development. Dev. Cell 5, 337–350
89. Khvorova, A., Reynolds, A. & Jayasena, S. D. Functional (2003).
siRNAs and miRNAs exhibit strand bias. Cell 115, 209–216 107. Sempere, L. F., Sokol, N. S., Dubrovsky, E. B., Berger,
(2003). E. M.& Ambros, V. Temporal regulation of microRNA
90. Lee, Y. et al. The nuclear RNase III Drosha initiates expression in Drosophila melanogaster mediated by
microRNA processing. Nature 425, 415–419 (2003). hormonal signals and broad-Complex gene activity. Dev.
91. Basyuk, E., Suavet, F., Doglio, A., Bordonne, R. & Biol. 259, 9–18 (2003).
Bertrand, E. Human let-7 stem-loop precursors harbor 108. Sempere, L. F. et al. Expression profiling of mammalian
features of RNase III cleavage products. Nucleic Acids Res. microRNAs uncovers a subset of brain-expressed
31,6593–6597 (2003). microRNAs with possible roles in murine and human
92. Zhang, H., Kolb, F. A., Jaskiewicz, L., Westhof, E. & neuronal differentiation. Genome Biol. 5, R13 (2004).
Filipowicz, W. Single processing center models for human 109. Calin, G. A. et al. MicroRNA profiling reveals distinct
Dicer and bacterial RNase III. Cell 118, 57–68 (2004). signatures in B cell chronic lymphocytic leukemias. Proc.
93. Lund, E., Guttinger, S., Calado, A., Dahlberg, J. E. & Natl Acad. Sci. USA 101, 11755–11760 (2004).
Kutay, U. Nuclear export of microRNA precursors. Science 110. Liu, C. G. et al. An oligonucleotide microchip for
303, 95–98 (2004). genomewide microRNA profiling in human and mouse
94. Park, W. et al. CARPEL FACTORY, a Dicer homolog, tissues. Proc. Natl Acad. Sci. USA 101, 9740–9744 (2004).
and HEN1, a novel protein, act in microRNA metabolism in 111. Schmittgen, T. D., Jiang, J., Liu, Q. & Yang, L. A
Arabidopsis thaliana. Curr. Biol. 12, 1484–1495 (2002). highthroughput method to monitor the expression of
95. Xie, Z. et al. Genetic and functional diversification of microRNA precursors. Nucleic Acids Res. 32, e43 (2004).
small RNA pathways in plants. PLoS Biol. 2, E104 (2004). 112. Babak, T., Zhang, W., Morris, Q., Blencowe, B. J. &
96. Papp, I. et al. Evidence for nuclear processing of plant Hughes, T. R. Probing microRNAs with microarrays: tissue
micro RNA and short interfering RNA precursors. Plant specificity and functional inference. RNA 10, 1813–1819
Physiol.132, 1382–1390 (2003). (2004).
97. Kurihara, Y. & Watanabe, Y. Arabidopsis micro-RNA 113. Barad, O. et al. MicroRNA expression detected by
biogenesis through Dicer-like 1 protein functions. Proc. Natl oligonucleotide microarrays: system establishment and
Acad. Sci. USA 101, 12753–12758 (2004). expression profiling in human tissues. Genome Res. 14,
98. Bollman, K. M. et al. HASTY, the Arabidopsis ortholog 2486–2494 (2004).
of exportin 5/MSN5, regulates phase change and 114. Miska, E. A. et al. Microarray analysis of microRNA
morphogenesis. Development 130, 1493–1504 (2003). expression in the developing mammalian brain. Genome
99. Telfer, A. & Poethig, R. S. HASTY: a gene that regulates Biol. 5, R68 (2004).
the timing of shoot maturation in Arabidopsis thaliana. 115. Sun, Y. et al. Development of a micro-array to detect
Development 125, 1889–1898 (1998). human and mouse microRNAs and characterization of
100. Park, M. Y. et al. Nuclear processing and export of expression in human organs. Nucleic Acids Res. 32, e188
microRNAs in Arabidopsis. Proc. Natl Acad. Sci. USA 102, (2004).
3691–3696 (2005). 116. Johnson, S. M., Lin, S. Y. & Slack, F. J. The time of
101. Vazquez, F., Gasciolli, V., Crete, P. & Vaucheret, H. appearance of the C. elegans let-7 microRNA is
The nuclear dsRNA binding protein HYL1 is required for transcriptionally controlled utilizing a temporal regulatory
microRNA accumulation and plant development, but not element in its promoter. Dev. Biol. 259, 364–379 (2003).
posttranscriptional transgene silencing. Curr. Biol. 14, 346– 117. Sewer A, Paul N, Landgraf P, Aravin A, Pfeffer S,
351 (2004). Brownstein M, et al.: Identification of clustered microRNAs
102. Han, M. H., Goud, S., Song, L. & Fedoroff, N. The using an ab initio prediction method. BMC Bioinformatics
Arabidopsis double-stranded RNA-binding protein HYL1 2005, 6:267.

International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36


29
118. Nam JW, Shin KR, Han J, Lee Y, Kim VN, Zhang BT: elegans by blocking LIN-14 protein synthesis after the
Human micro-RNA prediction through a probabilistic co- initiation of translation. Dev. Biol. 216, 671–680.
learning model of sequence and structure. Nucl Acids Res 135. Seggerson, K. et al. (2002) Two genetic circuits repress
2005, 33:3570-3581. the Caenorhabditis elegans heterochronic gene lin-28 after
119. Xue C, Li F, He T, Liu GP, Li Y, Zhang X: translation initiation. Dev. Biol. 243, 215–225.
Classification of real and pseudo microRNA precursors using 136. Zhao, Y. et al. (2005) Serum response factor regulates a
local structure sequence features and support vector machine. muscle-specific microRNA that targets Hand2 during
BMC Bioinformatics2005, 6:310. cardiogenesis. Nature 436, 214–220
120. Krol J, Sobczak K, Wilczynska U, Drath M, Jasinska A, 137. O‘Donnell, K.A. et al. (2005) c-Myc-regulated
Kaczynska D, et al.: Structural Features of MicroRNA microRNAs modulate E2F1 expression. Nature 435, 839–843
(miRNA) Precursors and Their Relevance to miRNA 138. Saito, Y. et al. (2006) Specific activation of microRNA-
Biogenesis and Small Interfering RNA/Short Hairpin RNA 127 with downregulation of the proto-oncogene BCL6 by
Design. J Biol Chem 2004, 279:42230-42239. chromatin- modifying drugs in human cancer cells. Cancer
121. Rehmsmeier M, Steffen P, Hochsmann M, Giegerich R: Cell 9, 435–443
Fast and effective prediction of microRNA/target duplexes. 139. Chen, J.F. et al. (2006) The role of microRNA-1 and
RNA 2004,10:1507-1517. microRNA-133 in skeletal muscle proliferation and
122. Saetrom O, Snove O Jr, Saetrom P: Weighted sequence differentiation. Nat. Genet. 38, 228–233
motifs as an improved seeding step in microRNA target 140. Boutz, P.L. et al. (2007) MicroRNAs regulate the
prediction algorithms. RNA 2005, 11:995-1003. expression of the alternative splicing factor nPTB during
123. Robins H, Li Y, Padgett RW: Incorporating structure to muscle development. Genes Dev. 21, 71–84 .
predict microRNA targets. Proc Natl Acad Sci USA 2005, 141. Maroney, P.A. et al. (2006) Evidence that microRNAs
102:4006-4009. are associated with translating messenger RNAs in human
124. Llave, C., Xie, Z., Kasschau, K. D. & Carrington, J. cells. Nat. Struct. Mol. Biol. 13, 1102–1107.
C.Cleavage of Scarecrow-like mRNA targets directed by a 142. Nottrott, S. et al. (2006) Human let-7a miRNA blocks
class of Arabidopsis miRNA. Science 297, 2053–2056 protein production on actively translating polyribosomes.
(2002). Nat. Struct. Mol.Biol. 13, 1108–1114.
125. Palatnik, J. F. et al. Control of leaf morphogenesis by 143. Petersen, C.P. et al. (2006) Short RNAs repress
microRNAs. Nature 425, 257–263 (2003). translation after initiation in mammalian cells. Mol. Cell 21,
126. Tang, G., Reinhart, B. J., Bartel, D. P. & Zamore, P. 533–542
D.A biochemical framework for RNA silencing in 144. Pillai, R.S. et al. (2005) Inhibition of translational
plants.Genes Dev. 17, 49– 63 (2003). initiation by let-7 microRNA in human cells. Science 309,
127. Yekta, S., Shih, I. H. & Bartel, D. P. MicroRNA- 1573–1576
directed cleavage of HOXB8 mRNA. Science 304, 594–596 145. Humphreys, D.T. et al. (2005) MicroRNAs control
(2004). translation initiation by inhibiting eukaryotic initiation factor
128. Carmell, M. A., Xuan, Z., Zhang, M. Q. & Hannon, G. 4E/cap and poly(A+) tail function. Proc. Natl. Acad. Sci. U.
J.The Argonaute family: tentacles that reach into RNAi, S. A. 102, 16961–16966.
developmental control, stem cell maintenance, and 146. Lim, L.P. et al. (2005) Microarray analysis shows that
tumorigenesis. Genes Dev. 16, 2733–2742 (2002). some microRNAs downregulate large numbers of target
129. Lee, R. C., Feinbaum, R. L. & Ambros, V. The C. mRNAs. Nature 433, 769–773.
elegans heterochronic gene lin-4 encodes small RNAs with 147. Bagga, S. et al. (2005) Regulation by let-7 and lin-4
antisense complementarity to lin-14. Cell75, 843–854 (1993). miRNAs results in target mRNA degradation. Cell 122, 553–
130. Bagga, S. et al. Regulation by let-7 and lin-4 miRNAs 563.
results in target mRNA degradation. Cell 122,553–563 148. Rehwinkel, J. et al. (2006) Genome-wide analysis of
(2005). mRNAs regulated by Drosha and Argonaute proteins in
131. Yekta S, Shih Ih, Bartel DP: MicroRNA-Directed Drosophila
Cleavage of HOXB8 mRNA. Science 2004, 304:594-596. melanogaster. Mol. Cell. Biol. 26, 2965–2975.
132. Rehwinkel J, Behm-Ansmant I, Gatfiled D, Izaurralde E: 149. Behm-Ansmant, I. et al. (2006) mRNA degradation by
A crucial role for GW182 and the DCP1:DCP2 decapping miRNAs and GW182 requires both CCR4:NOT deadenylase
complex in miRNA-mediated gene silencing. RNA 2005, and DCP1:DCP2 decapping complexes. Genes Dev. 20,
11:1640-1647. 1885–1898.
133. Liu J, Rivas FV, Wohlschlegel J, Yates JR, Parker R, 150. Schmitter, D. et al. (2006) Effects of Dicer and
Hannon GJ: A role for the P-body component GW182 in Argonaute downregulation on mRNA levels in human
microRNA function. Nat Cell Biol 2005, 7:1261-1266 HEK293 cells. Nucleic Acids Res. 34, 4801–4815
134. Olsen, P.H. and Ambros, V. (1999) The lin-4 regulatory 151. Giraldez, A.J. et al. (2006) Zebrafish MiR-430 promotes
RNA controls developmental timing in Caenorhabditis deadenylation and clearance of maternal mRNAs. Science
312, 75–79
International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
30
152. Wu, L. et al. (2006) MicroRNAs direct rapid 171. Vella, M. C., Reinert, K. & Slack, F. J. Architecture of a
deadenylation of mRNA.Proc. Natl. Acad. Sci. U. S. A. 103, validated microRNA::target interaction. Chem. Biol.11,
4034–4039 1619–1623 (2004).
153. Mishima, Y. et al. (2006) Differential regulation of 172. Hobert, O. Common logic of transcription factor and
germline mRNAs in soma and germ cells by zebrafish miR- microRNA action. Trends Biochem. Sci. 29, 462–468 (2004).
430. Curr. Biol. 16, 2135–2142. 173. Lee, R. C., Feinbaum, R. L. & Ambros, V. The C.
154. Bhattacharyya, S.N. et al. (2006) Relief of microRNA- elegans eterochronic gene lin-4 encodes small RNAs with
mediated translational repression in human cells subjected to antisense complementarity to lin-14. Cell 75, 843–854
stress. Cell 125,1111–1124 (1993).
155. Khvorova, A., Reynolds, A. & Jayasena, S. 174. Wightman, B., Ha, I. & Ruvkun, G. Posttranscriptional
D.Functional siRNAs and miRNAs exhibit strand bias.Cell regulation of the heterochronic gene lin-14 by lin-4 mediates
115, 209–216 (2003). temporal pattern formation in C. elegans. Cell 75, 855–862
156. Schwarz, D. S. et al. Asymmetry in the assembly of the (1993).
RNAi enzyme complex. Cell 115, 199–208 (2003). 175. Moss, E. G., Lee, R. C. & Ambros, V. The cold shock
157. Doench, J. G. & Sharp, P. A. Specificity of microRNA domain protein LIN-28 controls developmental timing in C.
target selection in translational repression. Genes Dev.18, elegans and is regulated by the lin-4 RNA. Cell 88, 637–646
504–511 (2004). (1997).
158. Vella, M. C., Choi, E. Y., Lin, S. Y., Reinert, K. & 176. Reinhart, B.J. et al. The 21-nucleotide let-7 RNA
Slack, F. J. The C. elegans microRNA let-7 binds to regulates developmental timing in Caenorhabditis elegans.
imperfect let-7 complementary sites from the lin-41 3′ UTR. Nature 403, 901–906 (2000).
Genes Dev. 18, 132–137 (2004). 177. Lin, S. Y. et al. The C. elegans hunchback homolog,
159. Brennecke, J., Stark, A., Russell, R. B. & Cohen, S. M. hbl-1,controls temporal patterning and is a probable
Principles of microRNA-target recognition. PLoS Biol.3, e85 microRNA target. Dev. Cell 4, 639–650 (2003).
(2005). 178. Abrahante, J. E. et al. The Caenorhabditis elegans
160. Lim, L. P. et al. Microarray analysis shows that some hunchback-like gene lin-57/hbl-1 controls developmental
microRNAs downregulate large numbers of target mRNAs. time and is regulated by microRNAs. Dev. Cell 4, 625–637
Nature 433, 769– 773 (2005). (2003).
161. Lim, L. P. et al. The microRNAs of Caenorhabditis 179. Slack, F. J. et al. The lin-41 RBCC gene acts in the C.
elegans. Genes Dev. 17, 991–1008 (2003). elegansheterochronic pathway between the let-7 regulatory
162. Grosshans, H., Johnson, T., Reinert, K. L., Gerstein, RNA and the LIN-29 transcription factor. Mol. Cell 5, 659–
M.& Slack, F. J. The temporal patterning microRNA let-7 669 (2000).
regulates several transcription factors at the larval to adult 180. Johnston, R. J. & Hobert, O. A microRNA controlling
transition in C. elegans. Dev. Cell 8, 321–330 (2005). left/right neuronal asymmetry in Caenorhabditis elegans.
163. Krek, A. et al. Combinatorial microRNA target Nature 426, 845–849 (2003).
predictions. Nature Genet. 37, 495–500 (2005). 181. Cohen, S. M. Bantam encodes a developmentally
164. Rehmsmeier, M., Steffen, P., Hochsmann, M. & regulated microRNA that controls cell proliferation and
Giegerich, R. Fast and effective prediction of microRNA/ regulates the proapoptotic gene hid in Drosophila. Cell 113,
target duplexes. RNA 10, 1507–1517 (2004) 25–36 (2003).
165. John, B. et al. Human microRNA targets. PLoS Biol. 2, 182. Xu, P., Vernooy, S. Y., Guo, M. & Hay, B. A. The
e363 (2004). Drosophila microRNA mir-14 suppresses cell death and is
166. Kiriakidou, M. et al. A combined computational– required for normal fat metabolism. Curr. Biol. 13, 790–795
experimental approach predicts human microRNA targets. (2003).
Genes Dev.18, 1165– 1178 (2004). 183. Chen, C. Z., Li, L., Lodish, H. F. & Bartel, D. P.
167. Enright, A. J. et al. MicroRNA targets in MicroRNAs modulate hematopoietic lineage differentiation.
Drosophila.Genome Biol. 5, R1 (2003). Science 303, 83–86 (2004).
168. Rajewsky, N. & Socci, N. D. Computational 184. Yekta, S., Shih, I. H. & Bartel, D. P. MicroRNA-
identification of microRNA targets. Dev. Biol. 267,529–535 directed cleavage of HOXB8 mRNA. Science 304, 594–596
(2004). (2004).
169. Stark, A., Brennecke, J., Russell, R. B. & Cohen, S. 185. Tang, G., Reinhart, B. J., Bartel, D. P. & Zamore, P. D.
M.Identification of Drosophila microRNA targets. PLoS A biochemical framework for RNA silencing in plants. Genes
Biol. 1, E60 (2003). Dev. 17, 49–63 (2003).
170. Pasquinelli, A., Reinhart, B., Slack, F., Maller, B. & 186. Emery, J. F. et al. Radial patterning of Arabidopsis
Ruvkun, G. Conservation across animal phylogeny of the shoots by class III HD-ZIP and KANADI genes. Curr. Biol.
sequence and temporal regulation of the 21 nucleotide C. 13, 1768–1774 (2003).zzzzzzzzzzzzzzzzzzzzzzzzzzzzzzz
elegans let-7 heterochronic regulatory RNA. Nature 408, 86– 187. Chen, X. A MicroRNA as a translational repressor of
89 (2000).
International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
31
APETALA2 in Arabidopsis flower development. Science 303, 202. He L, Thomson JM, Hemann MT, Hernando-Monge E,
2022–2025 (2004). Mu D, Goodson S, Powers S, Cordon-Cardo C, Lowe SW,
188. Palatnik, J. F. et al. Control of leaf morphogenesis by Hannon GJ, Hammond SM (2005) A microRNA polycistron
microRNAs. Nature 425, 257–263 (2003). as a potential human oncogene. Nature 435:828–833
189. Llave, C., Xie, Z., Kasschau, K. D. & Carrington, J. C. 203. Matsubara H, Takeuchi T, Nishikawa E, Yanagisawa K,
Cleavage of Scarecrow-like mRNA targets directed by a class Hayashita Y,Ebi H, Yamada H, Suzuki M, Nagino M,
of Arabidopsis miRNA. Science 297, 2053–2056 (2002). Nimura Y, Osada H, Takahashi T (2007) Apoptosis induction
190. Juarez, M. T., Kui, J. S., Thomas, J., Heller, B. A. & by antisense oligonucleotides against miR-17-5p and miR-
Timmermans, M. C. microRNA-mediated repression of 20a in lung cancers overexpressing miR-17-92. Oncogene
rolled leaf1 specifies maize leaf polarity. Nature 428, 84–88 26:6099–6105.
(2004). 204. Sylvestre Y, De Guire V, Querido E, Mukhopadhyay
191. Brennecke J, Hipfner DR, Stark A, Russell RB, Cohen UK, Bourdeau V, Major F, Ferbeyre G, Chartrand P (2007)
SM (2003) bantam encodes a developmentally regulated An E2F/miR-20a autoregulatory feedback loop. J Biol Chem
microRNA that controls cell proliferation and regulates the 282:2135–2143.
proapoptotic gene hid in Drosophila. Cell 113:25–36 205. Dews M, Homayouni A, Yu D, Murphy D, Sevignani
192. Lee RC, Feinbaum RL, Ambros V (1993) The C. C,Wentzel E,Furth EE, LeeWM, Enders GH,Mendell JT,
elegans heterochronic gene lin-4 encodes small RNAs with Thomas- Tikhonenko A (2006) Augmentation of tumor
antisense complementarity to lin-14. Cell 75:843–854 angiogenesis by a Myc-activated microRNA cluster. Nat
193. Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Genet 38:1060–1065
Yendamuri S, Shimizu M, Rattan S, Bullrich F, Negrini M, 206. O‘Donnell KA, Wentzel EA, Zeller KI, Dang CV,
Croce CM (2004) Human microRNA genes are frequently Mendell JT (2005) c-Myc-regulated microRNAs modulate
located at fragile sites and genomic regions involved in E2F1 expression. Nature 435:839–843.
cancers. Proc Natl Acad Sci USA 101:2999–3004 207. Johnson SM, GrosshansH, Shingara J, ByromM, Jarvis
194. Calin GA, Croce CM (2006) MicroRNA signatures in R, Cheng A, Labourier E, Reinert KL, Brown D, Slack FJ
human cancers. Nat Rev Cancer 6:857–866 (2005) RAS is regulated by the let-7 microRNA family. Cell
195. Gaur A, Jewell DA, Liang Y, Ridzon D, Moore JH, 120:635–647
Chen C, Ambros VR, Israel MA (2007) Characterization of 208. Takamizawa J, Konishi H, Yanagisawa K, Tomida S,
microRNA expression levels and their biological correlates in Osada H, Endoh H, Harano T, Yatabe Y, Nagino M, Nimura
human cancer cell lines. Cancer Res 67:2456–246 Y, Mitsudomi T, Takahashi T (2004) Reduced expression of
196. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, the let-7 microRNAs in human lung cancers in association
Peck D,Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, with shortened postoperative survival. Cancer Res 64:3753–
Downing JR, Jacks T, Horvitz HR, Golub TR (2005) 3756.
MicroRNA expression profiles classify human cancers. 209. Calin, G. A. et al. Frequent deletions and down
Nature 435:834–838. regulation of micro-RNA genes miR15 and miR16 at 13q14
197. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, in chronic lymphocytic leukemia. Proc. NatlAcad. Sci. USA
Noch E, Aldler H, Rattan S, Keating M, Rai K, Rassenti L, 99, 15524–15529 (2002)
Kipps T, Negrini M, Bullrich F, Croce CM (2002) Frequent 210. Cimmino, A. et al. miR-15 and miR-16 induce apoptosis
deletions and down-regulation of micro- RNA genes miR15 by targeting BCL2. Proc. Natl Acad. Sci.USA 102, 13944–
and miR16 at 13q14 in chronic lymphocytic leukemia. Proc 13949 (2005).
Natl Acad Sci USA 99:15524–15529. 211. Iorio, M. V. et al. MicroRNA gene expression
198. He L, Thomson JM, Hemann MT, Hernando-Monge E, deregulation in human breast cancer. Cancer Res. 65, 7065–
Mu D,Goodson S, Powers S, Cordon-Cardo C, Lowe SW, 7070 (2005).
Hannon GJ, Hammond SM (2005) A microRNA polycistron 212. Michael, M. Z., O‘Connor, S. M., van Holst Pellekaan,
as a potential human oncogene. Nature 435:828–833. N. G., Young, G. P. & James, R. J. Reduced accumulation of
199. O‘Donnell KA, Wentzel EA, Zeller KI, Dang CV, specific microRNAs in colorectal neoplasia. Mol. Cancer
Mendell JT (2005) c-Myc-regulated microRNAs modulate Res.1, 882–891 (2003).
E2F1expression. Nature 435:839–843. 213. Ciafre, S. A. et al. Extensive modulation of a set of
200. Johnson SM, GrosshansH, Shingara J, ByromM, Jarvis microRNAs in primary glioblastoma. Biochem.Biophys. Res.
R, Cheng A, Labourier E, Reinert KL, Brown D, Slack FJ Commun. 334, 1351–1358 (2005).
RAS is regulated by the let-7 microRNA family. Cell 214. Chan, J. A., Krichevsky, A. M. & Kosik, K. S.
120:635–647(2005). MicroRNA-21 is an antiapoptotic factor in human
201. Ota A, Tagawa H, Karnan S, Tsuzuki S, Karpas A, Kira glioblastoma cells. Cancer Res. 65, 6029–6033 (2005)
S,Yoshida Y, Seto M (2004) Identification and 215. Takamizawa, J. et al. Reduced expression of the let-
characterization of a novel gene, C13orf25, as a target for 7microRNAs in human lung cancers in association with
13q31-q32 amplification in malignant lymphoma. Cancer Res shortened postoperative survival. Cancer Res. 64, 3753–3756
64:3087–3095. (2004).
International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
32
216. Johnson, S. M. et al. RAS is regulated by the let- 232. Lujambio A, Ropero S, Ballestar E, Fraga MF, Cerrato
7microRNA family. Cell 120, 635–647 (2005). C, Setien F, Casado S, Suarez-GauthierA, Sanchez-
217. Lagos-Quintana, M. et al. Identification of tissuespecific CespedesM,Gitt A, Spiteri I, Das PP, Caldas C, Miska E,
microRNAs from mouse. Curr. Biol. 12,735–739 (2002). EstellerM (2007) Genetic unmasking of an epigenetically
218. Metzler, M., Wilda, M., Busch, K., Viehmann, S. & silenced microRNA in human cancer cells. Cancer Res
Borkhardt, A. High expression of precursor microRNA 67:1424–1429.
155/BIC RNA in children with Burkitt lymphoma.Genes 233. Saito Y, Liang G, Egger G, Friedman JM, Chuang JC,
Chromosomes Cancer 39, 167–169 (2004). Coetzee GA, Jones PA (2006) Specific activation of
219. Eis, P. S. et al. Accumulation of miR-155 and BIC RNA microRNA-127 with downregulation of the proto-oncogene
in human B cell lymphomas. Proc. Natl Acad. Sci. USA 102, BCL6 by chromatin modifying drugs in human cancer cells.
3627–3632 (2005). Cancer Cell 9:435–443.
220. Kluiver, J. et al. BIC and miR-155 are highly expressed 234. Mayr C, Hemann MT, Bartel DP (2007) Disrupting the
in Hodgkin, primary mediastinal and diffuse large B cell pairing between let-7 and Hmga2 enhances oncogenic
lymphomas. J. Pathol. 207, 243–249 (2005). transformation. Science 315:1576–1579
221. van den Berg, A. et al. High expression of B-cell 235. Lee YS, Dutta A (2007) The tumor suppressor
receptor inducible gene BIC in all subtypes of Hodgkin microRNA let-7 represses the HMGA2 oncogene. Genes Dev
lymphoma. Genes Chromosomes Cancer 37, 20–28 (2003). 21:1025–1030.236. Thomson JM, Newman M, Parker JS,
222. He, L. et al. A microRNA polycistron as a potential Morin-Kensicki EM, Wright T, Hammond SM (2006)
human oncogene. Nature 435, 828–833 (2005). Extensive post- transcriptional regulation of microRNAs and
223. O‘Donnell, K. A., Wentzel, E. A., Zeller, K. I., Dang, C. its implications for cancer. Genes Dev 20:2202–2207.
V.& Mendell, J. T. c-Myc-regulated microRNAs modulate 237. A. Orlacchio, G. Bernardi, A. Orlacchio, and S. Martino,
E2F1 expression. Nature 435, 839–843 (2005). ―Patented therapeutic RNAi strategies for neurodegenerative
224. Chang TC,Wentzel EA, Kent OA, Ramachandran diseases of the CNS,‖ Expert Opinion on Therapeutic
K,MullendoreM, Lee KH, Feldmann G, Yamakuchi M, Patents, vol. 18, no. 10, pp. 1161–1174, 2008.
Ferlito M, Lowenstein CJ, Arking DE, Beer MA, Maitra A, 238. N. Bushati and S. M. Cohen, ―microRNAs in neuro-
Mendell JT (2007) Transactivation of miR-34a by p53 degeneration,‖Current Opinion in Neurobiology, vol. 18, no.
broadly influences gene expression and promotes apoptosis. 3, pp.292–296, 2008.
Mol Cell 26:745–752 239. C. Patterson, J. W. Feightner, A. Garcia, G.-Y. R.
225. He L, He X, Lim LP, de Stanchina E, Xuan Z, Liang Y, Hsiung, C. MacKnight, and A. D. Sadovnick, ―Diagnosis and
Xue W,Zender L, Magnus J, Ridzon D, Jackson AL, Linsley treatment of dementia: 1. Risk assessment and primary
PS, Chen C, Lowe SW, Cleary MA, Hannon GJ (2007) A prevention of Alzheimer disease,‖ Canadian Medical
microRNA component of the p53 tumour suppressor Association Journal,vol.178, no.5, pp. 548–556, 2008.
network. Nature 447:1130–1134. 240. A. Orlacchio, G. Bernardi, A. Orlacchio, and S. Martino,
226. Ma L, Teruya-Feldstein J, Weinberg RA (2007) Tumour ―RNA interference as a tool for Alzheimer‘s disease
invasion and metastasis initiated by microRNA-10b in breast therapy,‖Mini-Reviews in Medicinal Chemistry, vol. 7, no.
cancer. Nature 449:682–688. 11, pp.1166–1176, 2007.
227. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, 241. E. Costanzi, S. Martino, E. Persichetti, et al., ―Effects of
Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, vitamin C on fibroblasts from sporadic Alzheimer‘s disease
Downing JR, Jacks T, Horvitz HR, Golub TR (2005) patients,‖ Neurochemical Research, vol. 33, no. 12, pp.
MicroRNA expression profiles classify human cancers. 2510–2515, 2008.
Nature 435:834–838. 242. E. A. Rogaeva, S. Premkumar, J. Grubber, et al., ―An α-
228. Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter 2-macroglobulin insertion-deletion polymorphism in
JM, Castle J, Bartel DP, Linsley PS, Johnson JM (2005) Alzheimer disease,‖ Nature Genetics, vol. 22, no. 1, pp. 19–
Microarray analysis shows that some microRNAs 22,1999.
downregulate large numbers of target mRNAs. Nature 243. J. W. Freeman, M. Simmons, and E. Matos, ―Parkinson
433:769–773. disease: an incremental challenge,‖ South Dakota Medicine,
229. Tang F, Hajkova P, Barton SC, Lao K, Surani MA vol. 60, no. 12, pp. 481–484, 2007.
(2006) MicroRNA expression profiling of single whole 244. C. Balducci, L. Pierguidi, E. Persichetti, et al.,
embryonic stem cells. Nucleic Acids Res 34:e9. ―Lysosomal hydrolases in cerebrospinal fluid from subjects
230. He L, Thomson JM, Hemann MT, Hernando-Monge E, with Parkinson‘s disease,‖ Movement Disorders, vol. 22, no.
Mu D, Goodson S, Powers S, Cordon-Cardo C, Lowe SW, 10, pp. 1481–1484, 2007.
Hannon GJ,Hammond SM (2005) A microRNA polycistron 245. W. Stamey and J. Jankovic, ―Impulse control disorders
as a potential human oncogene. Nature 435:828–833. and pathological gambling in patients with parkinson
231. Chen CZ, Li L, Lodish HF, Bartel DP (2004) disease,‖Neurologist, vol. 14, no. 2, pp. 89–99, 2008.
MicroRNAs modulate hematopoietic lineage differentiation.
Science 303:83–86.
International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
33
246. G. G. Kovacs and H. Budka, ―Prion diseases: from 260. W. J. Lukiw, ―Micro-RNA speciation in fetal, adult and
protein to cell pathology,‖ American Journal of Pathology, Alzheimer‘s disease hippocampus,‖ NeuroReport, vol. 18,
vol. 172, no. 3, pp. 555–565, 2008. no. 3, pp. 297–300, 2007.
247. V. B. Tripathi and A. Al-Chalabi, ―Molecular insights 261. V. Boissonneault, I. Plante, S. Rivest, and P. Provost,
and therapeutic targets in amyotrophic lateral sclerosis,‖ CNS ―microRNA-298 and microRNA-328 regulate expression of
and Neurological Disorders—Drug Targets, vol. 7, no. 1, pp. mouse β-amyloid precursor protein-converting enzyme
11–19, 2008. 1,‖Journal of Biological Chemistry, vol. 284, no. 4, pp.
248. A. Orlacchio, G. Bernardi, A. Orlacchio, and S. Martino, 1971– 981, 2009.
―Genetics of amyotrophic lateral sclerosis,‖ Recent Research 262. W. J. Lukiw, Y. Zhao, and J. G. Cui, ―An NF-
in Developmental Neuroscience, pp. 17–54, 2007. κBsensitive micro RNA-146a-mediated inflammatory circuit
249. A. Orlacchio, T. Kawarai, A. Totaro, et al., ―Hereditary in alzheimer disease and in stressed human brain cells,‖
spastic paraplegia: clinical genetic study of 15 families,‖ Journalof Biological Chemistry, vol. 283, no. 46, pp. 31315–
Archives of Neurology, vol. 61, no. 6, pp. 849–855, 2004. 31322, 2008.
250. A. Orlacchio, T. Kawarai, F. Gaudiello, P. H. St. 263. S.-L. Ang, ―Transcriptional control of midbrain
George-Hyslop, R. Floris, and G. Bernardi, ―New locus for dopaminergic neuron development,‖ Development, vol. 133,
hereditary spastic paraplegia maps to chromosome 1p31.1– no. 18, pp. 3499–3506, 2006.
1p21.1,‖Annals of Neurology, vol. 58, no. 3, pp. 423–429, 264. Y. Lee, R. C. Samaco, J. R. Gatchel, C. Thaller, H. T.
2005. Orr, and H. Y. Zoghbi, ―miR-19, miR-101 and miR-130 co-
251. E. C. Stack and R. J. Ferrante, ―Huntington‘s disease: regulate ATXN1 levels to potentially modulate SCA1
progress and potential in the field,‖ Expert Opinion on pathogenesis,‖Nature Neuroscience, vol. 11, no. 10, pp.
Investigational Drugs, vol. 16, no. 12, pp. 1933–1953, 2007. 1137–1139, 2008.
252. E. J. Wild and S. J. Tabrizi, ―Huntington‘s disease 265. A. N. Packer, Y. Xing, S. Q. Harper, L. Jones, and B. L.
phenocopy syndromes,‖ Current Opinion in Neurology, vol. Davidson, ―The bifunctional microRNA miR-9/miR-9∗
20, no. 6, pp. 681–687, 2007. regulates REST and CoREST and is downregulated in
253. S. Martino, C. Emiliani, B. Tancini, et al., ―Absence of Huntington‘s disease,‖ Journal of Neuroscience, vol. 28, no.
metabolic cross-correction in Tay-Sachs cells: implications 53, pp. 14341–14346, 2008.
for gene therapy,‖ Journal of Biological Chemistry, vol. 266. R. Johnson, C. Zuccato, N. D. Belyaev, D. J. Guest, E.
277,no. 23, pp. 20177–20184, 2002. Cattaneo, and N. J. Buckley, ―A microRNA-based gene
254. S. Martino, C. Cavalieri, C. Emiliani, et al., ―Restoration dysregulation pathway in Huntington‘s disease,‖
of the GM2 ganglioside metabolism in bone marrowderived Neurobiology of Disease, vol. 29, no. 3, pp. 438–445, 2008.
stromal cells from Tay-Sachs disease animal 267. A. Clop, F. Marcq, H. Takeda, et al., ―A mutation
model,‖Neurochemical Research, vol. 27, no. 7-8, pp. 793– creating a potential illegitimate microRNA target site in the
800, 2002. myostatin gene affects muscularity in sheep,‖ Nature
255. S. Martino, P. Marconi, B. Tancini, et al., ―A direct gene Genetics, vol. 38,no. 7, pp. 813–818, 2006.
transfer strategy via brain internal capsule reverses 268. J. A. Chan, A. M. Krichevsky, and K. S. Kosik,
thebiochemical defect in Tay-Sachs disease,‖ Human ―microRNA- 21 is an antiapoptotic factor in human
MolecularGenetics, vol. 14, no. 15, pp. 2113–2123, 2005. glioblastoma cells,‖ Cancer Research, vol. 65, no. 14, pp.
256. S. Martino and A. Orlacchio, ―Cellule staminali e GM2 6029–6033, 2005.
gangliosidosi,‖ Annali dell’Istituto Superiore di Sanit`a, pp. 269. J. Silber, D. A. Lim, C. Petritsch, et al., ―miR-124 and
27–28, 2005. miR-137 inhibit proliferation of glioblastoma multiforme
257. S. Martino, I. di Girolamo, C. Cavazzin, et al., ―Neural cells and induce differentiation of brain tumor stem cells,‖
precursor cell cultures from GM2 gangliosidosis animal BMCMedicine, vol. 6, article 14, 2008.
models recapitulate the biochemical and molecular hallmarks 270. J. Pierson, B. Hostager, R. Fan, and R. Vibhakar,
of the brain pathology,‖ Journal of Neurochemistry, vol. 109, ―Regulation of cyclin dependent kinase 6 by microRNA 124
no. 1, pp. 135–147, 2009. in medulloblastoma,‖Journal of Neuro-Oncology, vol. 90,
258. M. S. Weinberg and M. J. A. Wood, ―Short non-coding no. 1, pp. 1–7, 2008.
RNA biology and neurodegenerative disorders: novel disease 271. J. S.Wei, Y. K. Song, S. Durinck, et al., ―The MYCN
targets and therapeutics,‖ HumanMolecular Genetics, vol. oncogene is a direct target of miR-34a,‖ Oncogene, vol. 27,
18,no. R1, pp. R27–R39, 2009. no. 39, 5204–5213, 2008.
259. S. S. H´ebert, K. Horr´e, L. Nicola¨ı, et al., ―Loss of 272. Y. Chen and R. L. Stallings, ―Differential patterns of
microRNA cluster miR-29a/b-1 in sporadic Alzheimer‘s microRNA expression in neuroblastoma are correlated with
disease correlates with increased BACE1/β-secretase prognosis, differentiation, and apoptosis,‖ Cancer Research,
expression,‖ Proceedings of the National Academy of vol. 67, no. 3, pp. 976–983, 2007.
Sciences of the United States of America, vol. 105, no. 17, 273. A. Bottoni, D. Piccin, F. Tagliati, A. Luchin, M. C.
pp. 6415–6420, 2008. Zatelli, and E. C. D. Uberti, ―miR-15a and miR-16-1 down-

International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36


34
regulation in pituitary adenomas,‖ Journal of Cellular 289. M. M. Hoehn and M. D. Yahr, ―Parkinsonism: onset,
Physiology, vol. 204, no. 1, pp. 280–285, 2005. progression, and mortality,‖ Neurology, vol. 57, no. 10, pp.
274. A. P. Heaney, ―Pituitary tumour pathogenesis,‖ British S11–S26, 2001.
MedicalBulletin, vol. 75-76, no. 1, pp. 81–97, 2005.. 290. R. L. Nussbaum and M. H. Polymeropoulos, ―Genetics
275. A. Cimmino, G. A. Calin, M. Fabbri, et al., ―miR-15 and of Parkinson‘s disease,‖ Human Molecular Genetics, vol. 6,
miR-16 induce apoptosis by targeting BCL2,‖ Proceedings of no. 10, pp. 1687–1691, 1997.
the National Academy of Sciences of the United States of 291. M. H. Polymeropoulos, C. Lavedan, E. Leroy, et al.,
America,vol. 102, no. 39, pp. 13944–13949, 2005. ―Mutation in the α-synuclein gene identified in families with
276. S. A. Ciafr`e, S. Galardi, A. Mangiola, et al., ―Extensive Parkinson‘s disease,‖ Science, vol. 276, no. 5321, pp. 2045–
modulation of a set of microRNAs in primary glioblastoma,‖ 2047, 1997.
Biochemical and Biophysical Research Communications, 292. V. M. Miller, H. Xia, G. L. Marrs, et al., ―Allele-specific
vol.334, no. 4, pp. 1351–1358, 2005. silencing of dominant disease genes,‖ Proceedings of the
277. T. Papagiannakopoulos and K. S. Kosik, ―microRNAs: National Academy of Sciences of the United States of
regulators of oncogenesis and stemness,‖ BMC Medicine, vol. America, vol. 100, no. 12, pp. 7195–7200, 2003.
6, article 15, 2008. 293. P. Gonzalez-Alegre, V. M. Miller, B. L. Davidson, and
278. S. Lawler and E. A. Chiocca, ―Emerging functions of H. L. Paulson, ―Toward therapy for DYT1 dystonia: allele-
microRNAs in glioblastoma,‖ Journal of Neuro-Oncology, specific silencing of mutant Torsin A,‖ Annals of Neurology,
vol. 92, no. 3, pp. 297–306, 2009. vol. 53,no. 6, pp. 781–787, 2003
279. E. Ferretti, E. De Smaele, A. Po, et al., ―microRNA 294. J. Kim, K. Inoue, J. Ishii, et al., ―A microRNA feedback
profiling in humanmedulloblastoma,‖ International Journal circuit in midbrain dopamine neurons,‖ Science, vol. 317, no.
of Cancer, vol. 124, no. 3, pp. 568–577, 2009. 5842, pp. 1220–1224, 2007.
280. M. Goedert and M. G. Spillantini, ―A century of 295. P. van den Munckhof, K. C. Luk, L. Ste-Marie, et al.,
Alzheimer‘s disease,‖ Science, vol. 314, no. 5800, pp. 777– ―Pitx3 is required for motor activity and for survival of a
781, 2006. subset of midbrain dopaminergic neurons,‖ Development,
281. P. T. Lansbury and H. A. Lashuel, ―A century-old vol. 130, no.11, pp. 2535–2542, 2003
debate on protein aggregation and neurodegeneration enters 296. J. Fuchs, J. C. Mueller, P. Lichtner, et al., ―The
the clinic,‖ Nature, vol. 443, no. 7113, pp. 774–779, 2006. transcription factor PITX3 is associated with sporadic
282. R. S. Turner, ―Alzheimer‘s disease,‖ Seminars in Parkinson‘s disease,‖ Neurobiology of Aging, vol. 30, no. 5,
Neurology, vol. 26, no. 5, pp. 499–506, 2006. pp. 731–738, 2009.
283. Y. Ohyagi and T. Tabira, ―Intracellular amyloid β- 297. T. L. Cuellar, T. H. Davis, P. T. Nelson, et al., ―Dicer
protein and its associated molecules in the pathogenesis of loss in striatal neurons produces behavioral and
Alzheimer‘s disease,‖ Mini-Reviews in Medicinal Chemistry, neuroanatomical phenotypes in the absence of
vol. 6, no. 10, pp. 1075–1080, 2006. neurodegeneration,‖ Proceedings of the National Academy of
284. D. Goldgaber, M. I. Lerman, and O.W.McBride, Sciences of the United States of America, vol. 105, no. 14, pp.
―Characterization and chromosomal localization of a cDNA 5614–5619, 2008.
encodingbrain amyloid of Alzheimer‘s disease,‖ Science, 298. A. H. Evans and A. J. Lees, ―Dopamine dysregulation
vol. 235, no.4791, pp. 877–880, 1987. syndrome in Parkinson‘s disease,‖ Current Opinion in
285. C. E. Hunt and A. J. Turner, ―Cell biology, regulation Neurology, vol. 17, no. 4, pp. 393–398, 2004.
and inhibition of β-secretase (BACE-1),‖ FEBS Journal, vol. 299. G. Wang, J. M. van der Walt, G. Mayhew, et al.,
276, no. 7, pp. 1845–1859, 2009. ―Variation in the miRNA-433 binding site of FGF20 confers
286. W.-X. Wang, B. W. Rajeev, A. J. Stromberg, et al., ―The Risk for Parkinson disease by overexpression of α-
expression of microRNA miR-107 decreases early in synuclein,‖ American Journal of Human Genetics, vol. 82,
Alzheimer‘s disease and may accelerate disease progression no. 2, pp. 283–289, 2008.
through regulation of β-site amyloid precursor 300. K. Nakamura and M. J. Aminoff, ―Huntington‘s disease:
proteincleaving enzyme 1,‖ Journal of Neuroscience, vol. 28, clinical characteristics, pathogenesis and therapies,‖ Drugs of
no. 5, pp.1213–1223, 2008. Today, vol. 43, no. 2, pp. 97–116, 2007.
287. S. Bicker and G. Schratt, ―microRNAs: tiny regulators 301. L. Zhang, J. Huang, N. Yang, et al., ―microRNAs
of synapse function in development and disease,‖ Journal of exhibit high frequency genomic alterations in human cancer,‖
Cellular and Molecular Medicine, vol. 12, no. 5A, pp. 1466– Proceedings of the National Academy of Sciences of the
1476, 2008. United States of America, vol. 103, no. 24, pp. 9136–9141,
288. D. C. Carrettiero, I. Hernandez, P. Neveu, T. Papa- 2006.
giannakopoulos, and K. S. Kosik, ―The cochaperone BAG2 302. A. Esquela-Kerscher and F. J. Slack, ―Oncomirs—
sweeps paired helical filament-insoluble tau from the microRNAs with a role in cancer,‖ Nature Reviews Cancer,
microtubule,‖Journal of Neuroscience, vol. 29, no. 7, pp. vol. 6, no. 4, pp. 259–269, 2006.
2151–2161, 2009. 303. T. Papagiannakopoulos, A. Shapiro, and K. S. Kosik,
―microRNA-21 targets a network of key tumor-suppressive
International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
35
pathways in glioblastoma cells,‖ Cancer Research, vol. 68, in vivo with ‗antagomirs‘. Nature 438:685–689.
no.19, pp. 8164–8172, 2008. 319. Ebert MS, Neilson JR, Sharp PA (2007) MicroRNA
304. M. S. Nicoloso and G. A. Calin, ―microRNA sponges: competitive inhibitors of small RNAs in mammalian
involvement in brain tumors: from bench to bedside,‖ Brain cells. NatMethods 4:721–726
Pathology, vol.18, no. 1, pp. 122–129, 2008. 320. Tsuda N, Kawano K, Efferson CL, Ioannides CG (2005)
305. G. A. Calin, C. Sevignani, C. D. Dumitru, et al., Synthetic microRNA and double-stranded RNA targeting the
―Human microRNA genes are frequently located at fragile 3′- untranslated region of HER-2/neu mRNA inhibit HER-2
sites and genomic regions involved in cancers,‖ Proceedings protein expression in ovarian cancer cells. Int J Oncol
of the National Academy of Sciences of the United States of 27:1299–1306.
America, vol. 101, no. 9, pp. 2999–3004, 2004. 321. Yeung ML, Bennasser Y, Myers T, Jiang G, Benkirane
306. S. Volinia, G. A. Calin, C.-G. Liu, et al., ―A microRNA M, Jeang KT:Changes in microRNA expression profiles in
expression signature of human solid tumors defines cancer HIV-1-transfected human cells. Retrovirology 2005, 2:81.
gene targets,‖ Proceedings of the National Academy of 322. Cai X, Schafer A, Lu S, Bilello JP, Desrosiers RC,
Sciencesof the United States of America, vol. 103, no. 7, pp. Edwards R, et al.:Epstein-Barr virus microRNAs are
2257–2261, 2006. evolutionarily conserved and differentially expressed. PLoS
307. L. He, J. M. Thomson, M. T. Hemann, et al., ―A Pathog 2006, 2:e23.
microRNA polycistron as a potential human oncogene,‖ 323. Hariharan M, Scaria V, Pillai B, Brahmachari SK:
Nature, vol. 435, no. 7043, pp. 828–833, 2005. Targets for human encoded microRNAs in HIV genes.
308. G. A. Calin and C. M. Croce, ―microRNA signatures in Biochem Biophys Res Commun 2005, 337:1214-1218.
human cancers,‖ Nature Reviews Cancer, vol. 6, no. 11, pp. 324. Weinberg MS, Morris KV: Are Viral-Encoded
857–866, 2006. MicroRNAs Mediating Latent HIV-1 Infection? DNA and
309. Lecellier CH, Dunoyer P, Arar K, Lehmann-Che J, Cell Biology 2006, 25:223-231
Eyquem S, Himber C, et al.: A Cellular MicroRNA Mediates 325. Sevignani C, Calin G, Siracusa L, Croce C: Mammalian
Antiviral Defense in Human Cells. Science 2005, 308:557- microRNAs: a small world for fine-tuning gene expression.
560. Mammalian Genome 2006, 17:189-202.
310. Calin GA, Liu CG, Sevignani C, Ferracin M, Felli N, 326. Cummins JM, He Y, Leary RJ, Pagliarini R, Diaz LA Jr,
Dumitru CD, etal.: MicroRNA profiling reveals distinct Sjoblom T, et al.: The colorectal microRNAome. Proc Natl
signatures in B cell chronic lymphocytic leukemias. Proc Acad Sci USA 2006, 103:3687-3692.
Natl Acad Sci USA 2004, 101:11755-11760. 327. Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A,
311. Bennasser Y, Le SY, Yeung ML, Jeang KT: HIV-1 Petrocca F, et al.: A microRNA expression signature of
encoded candidate micro-RNAs and their cellular targets. human solid tumors defines cancer gene targets. Proc Natl
Retrovirology 2004, 1:43. Acad Sci USA 2006, 103:2257-2261.
312. Yeung ML, Bennasser Y, Myers T, Jiang G, Benkirane 328. Calin GA, Ferracin M, Cimmino A, Di Leva G, Shimizu
M, Jeang KT: Changes in microRNA expression profiles in M, Wojcik SE, et al.: A MicroRNA Signature Associated
HIV-1-transfected human cells. Retrovirology 2005, 2:81. with Prognosis and Progression in Chronic Lymphocytic
313. Jopling CL, Yi M, Lancaster AM, Lemon SM, Sarnow Leukemia. N Engl J Med 2005, 353:1793-1801.
P: Modulation of Hepatitis C Virus RNA Abundance by a 329. Krutzfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl
Liver-Specific MicroRNA. Science 2005, 309:1577-1581. T, Manoharan M, et al.: Silencing of microRNAs in vivo
314. Lu J, Getz G, Miska EA, varez-Saavedra E, Lamb J, with/'antagomirs/'.Nature 2005, 438:685-689.
Peck D, et al.:MicroRNA expression profiles classify human 330. Weiler J, Hunziker J, Hall J: Anti-miRNA
cancers. Nature 2005, 435:834-838. oligonucleotides (AMOs): ammunition to target miRNAs
315. Gabriel G, Dauber B, Wolff T, Planz O, Klenk HD, implicated in human disease? Gene Ther 2005, 13:496-502.
Stech J: The viral polymerase mediates adaptation of an avian 331. Vester B, Wengel J: LNA (locked nucleic acid): high-
influenza virus to a mammalian host. Proc Natl Acad Sci affinity targeting of complementary RNA and DNA.
USA 2005, 102:18590-18595. Biochemistry 2004, 43:13233-13241.
316. Hutvagner G, Simard MJ, Mello CC, Zamore PD (2004) 332. Castoldi M, Schmidt S, Benes V, Noerholm M, Kulozik
Sequence-specific inhibition of small RNA function. PLoS AE, Hentze MW, et al.: A sensitive array for microRNA
Biol 2:E98. expression profiling (miChip) based on locked nucleic acids
317. Meister G, Landthaler M, Dorsett Y, Tuschl T (2004) (LNA). RNA 2006,12:913-920.
Sequencespecific inhibition of microRNA- and siRNA- 333. Schwab R, Ossowski S, Riester M, Warthmann N,
induced RNA silencing. RNA 10:544–550. Weigel D: Highly Specific Gene Silencing by Artificial
318. Krutzfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl MicroRNAs in Arabidopsis. Plant Cell 2006, 18:1121-1133.
T, Manoharan M, Stoffel M (2005) Silencing of microRNAs

Source of support: Nil; Conflict of interest: None declared


International Journal of Research in Biotechnology and Biochemistry 2011; 1(1) : 11-36
36

View publication stats

You might also like