Micrornas: Regulators of Gene Expression and Cell Differentiation

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

From www.bloodjournal.org by guest on March 11, 2018. For personal use only.

Review in translational hematology

MicroRNAs: regulators of gene expression and cell differentiation


Ramesh A. Shivdasani

The existence and roles of a class of transcript, often one encoding a transcrip- level expression of unwanted genes and
abundant regulatory RNA molecules have tion factor, that controls a specific aspect hence refine unilineage gene expression,
recently come into sharp focus. Micro- of development, cell differentiation, or may be especially amenable to evaluation
RNAs (miRNAs) are small (approximately physiology. However, computational algo- in models of hematopoiesis. This review
22 bases), non–protein-coding RNAs that rithms predict up to hundreds of putative summarizes current understanding of
recognize target sequences of imperfect targets for individual miRNAs, single tran- miRNA mechanisms, outlines some of
complementarity in cognate mRNAs and scripts may be regulated by multiple mi- the important outstanding questions, and
either destabilize them or inhibit protein RNAs, and miRNAs may either eliminate describes studies that attempt to define
translation. Although mechanisms of target gene expression or serve to fine- miRNA functions in hematopoiesis.
miRNA biogenesis have been elucidated tune transcript and protein levels. Theo- (Blood. 2006;108:3646-3653)
in some detail, there is limited apprecia- retical considerations and early experi-
tion of their biological functions. Re- mental results hence suggest diverse
ported examples typically focus on miRNA roles for miRNAs as a class. One appeal-
regulation of a single tissue-restricted ing possibility, that miRNAs eliminate low- © 2006 by The American Society of Hematology

miRNAs, the sounds of (gene) silence


MicroRNAs (miRNAs) are small, often phylogenetically con- respective targets, the protein-coding mRNA products of the lin-14
served, non–protein-coding RNAs that mediate posttranscriptional and lin-41 genes.5,6 Although posttranscriptional gene silencing by
gene repression by inhibiting protein translation or by destabilizing small regulatory RNAs was initially regarded as a biological
target transcripts. miRNAs recognize target sites, most commonly curiosity, perhaps restricted to certain lower species, we now know
found in the 3⬘-untranslated regions (UTRs) of cognate mRNAs, that many multicellular organisms express hundreds of miRNAs
through imperfect base-pairing, with 1 or more mismatches in that vary in abundance across cell and tissue types.7-10 miRNAs are
sequence complementarity. miRNAs are believed to fine-regulate a found in multicellular plant and animal species but not in unicellu-
diverse array of biological processes, and a convergence of genetic, lar organisms such as yeast. By definition, miRNAs correspond to
biochemical, and structural studies has led to rapid growth in cloned small RNAs that, like the lin-4 and let-7 products, are
understanding of their synthesis and molecular mechanisms. In processed from stem-loop precursors transcribed from genes other
contrast, the precise biological functions of these approximately than the ones they regulate. Endogenous small RNAs that do not
22-base RNA products are less clear. To date, only a handful of meet these criteria and are known collectively as small-interfering
studies provide definitive evidence of a role for a specific miRNA RNAs (siRNAs) derive from long double-stranded RNA (dsRNA)
in vertebrate biology, although such roles are probably pervasive. precursors that in some species may be transcribed from repeat
The limits on current appreciation simply reflect the infancy of the elements, heterochromatin, or transposons.3 The 2 classes of short
field, and it is safe to predict a vast growth in functional studies that regulatory RNAs differ mainly in their origins and not in their
will refine understanding of the roles of individual miRNAs. functions,11 which may be grouped under the single moniker of
The history and general features of miRNAs have been RNA interference (RNAi), although siRNA sequences are usually
reviewed extensively in a number of different contexts.1-3 Here I perfectly complementary to those of their RNA targets (Figure 1).
review the biogenesis and mechanisms of miRNAs briefly before
exploring the current appreciation of the role of miRNAs in cell
differentiation, with emphasis on hematopoiesis. Many of the Salient concepts in miRNA function
general and biochemical insights derive from experiments con-
ducted in worms and flies, and these studies are discussed when In zebrafish development, where miRNA expression dynamics are
appropriate. Although investigation in invertebrate species also the best characterized to date, most miRNAs appear relatively late
provides many functional insights,4 this review will focus on in embryogenesis and reveal tissue-specific distributions and
miRNA functions in vertebrate animals. requirements.12-14 In certain mammalian organs, including bone
The first miRNAs, lin-4 and let-7, were each identified, some 7 marrow, a handful of tissue-specific miRNAs tend to dominate the
years apart, on the basis of their mutant phenotypes in forward miRNA expression profile,10,15 and in a limited analysis of mouse
genetic screens in the worm Caenorhabditis elegans. Both entities miRNAs, expression profiles differed between fetal and adult
control developmental timing by repressing translation of their forms of the same tissue.16 Taken together with the original

From the Department of Medical Oncology, Dana-Farber Cancer Institute, as Blood First Edition Paper, August 1, 2006; DOI 10.1182/blood-2006-
Boston, MA; and the Departments of Medicine, Brigham & Women’s Hospital 01-030015.
and Harvard Medical School, Boston, MA.

Submitted January 30, 2006; accepted July 18, 2006. Prepublished online © 2006 by The American Society of Hematology

3646 BLOOD, 1 DECEMBER 2006 䡠 VOLUME 108, NUMBER 12


From www.bloodjournal.org by guest on March 11, 2018. For personal use only.

BLOOD, 1 DECEMBER 2006 䡠 VOLUME 108, NUMBER 12 miRNA REGULATION OF HEMATOPOIESIS 3647

Figure 1. miRNAs and siRNAs: differences in biogenesis and


properties. siRNAs (left) derive from long endogenous dsRNA
molecules that form either long hairpins or bimolecular duplexes.
Processing of these dsRNA precursors can generate many
different siRNAs from both strands. In contrast, processing of the
shorter hairpin structures known as pre-miRNAs (right) produces
a single miRNA molecule from one arm of the hairpin precursor.
siRNAs recognize their target transcripts with perfect sequence
complementarity (left), whereas miRNAs typically have a limited
number of mismatches with their mRNA target sequences (right).
Both classes of small regulatory RNA molecules cause posttran-
scriptional silencing of protein-coding genes.

identification of worm miRNAs as regulators of developmental occur by intrinsic decay. This particular role may be especially
timing, such observations have led to the notion that a principal pertinent in hematopoiesis. In a process known as multilineage
function of miRNAs is to control cell differentiation and develop- gene priming, immature blood progenitors initially activate a leaky
ment. Indeed, mouse embryonic stem cells with an engineered and promiscuous transcriptional program; as cells commit to a
inability to synthesize miRNAs are viable but fail to differentiate, restricted fate, unilineage gene expression is reinforced and tran-
and zebrafish embryos that are unable to produce mature miRNAs scripts affiliated with alternative blood cell types disappear.25 It is
develop normally at first but display specific morphogenetic tempting to speculate that miRNAs drive the latter phase, an idea
defects during gastrulation and organogenesis.13,17 Specific and that is as yet untested.
prominent miRNA functions are illustrated in part by the roles of Animal genomes not only encode a large number of tissue-
miR-196 in vertebrate limb development,18 of the miR-1 cluster in restricted miRNAs, but these miRNAs can be expressed in high,
cardiomyocyte differentiation,19 and of miR-181 in maintaining a albeit variable, abundance, with estimates in C elegans ranging
balance between circulating T and B lymphocytes.15 However, from 800 to 50 000 molecules per cell.26 Coupled with the limited
miRNAs are expressed widely in developing and adult tissues and evolutionary constraints on small non–protein-coding genes,
their functions are likely to be widespread, diverse, and ranging miRNAs may be positioned ideally to regulate tissue-specific gene
from seminal to subtle depending on the context. expression. As the latter function lies at the heart of cell differentia-
In the best-studied examples in worms and mammals, miRNAs tion, it is fair to speculate that miRNAs as a class play a vital role in
control key developmental processes by inhibiting just one or a few generating and maintaining distinctive cell lineages in all multicel-
mRNA targets. As we shall see, however, the typical miRNA can lular species. However, forward genetic screens in C elegans and
potentially regulate dozens or scores of genes,20,21 and most targets Drosophila have yielded very few miRNAs, possibly reflecting the
contain isolated miRNA recognition sites that may be inadequate small size of the coding unit, subtlety in mutant phenotypes, and a
for complete gene silencing. These considerations prompted Bartel substantial degree of functional redundancy. Instead, current cata-
and Chen to propose the presence of selective pressure on many logs of miRNAs derive largely from the results of cDNA cloning.
genes to avoid undesirable interference from miRNAs that are both In May 2006, a unified database (miRBase)27 recognized 462 and
widespread and have limited complementarity requirements.22 340 individual miRNAs in the human and mouse, respectively.
Stark and colleagues recently reported that genes that seem to avoid Nomenclature and annotation for all species are assigned and
miRNA regulation (“antitargets”) tend to function in processes managed centrally, with the “miR” designation followed by a
common to all cells, whereas putative targets are more commonly unique identifying number.11,28 Identical miRNAs are given the
implicated in developmental processes, especially organogenesis.23 same number, regardless of organism, as are nearly identical
In several tested examples in Drosophila embryos, miRNAs and sequences that are presumed to represent orthologous miRNAs. Highly
some predicted targets are indeed expressed in nonoverlapping similar miRNAs within a species also are assigned the same
adjacent distributions, whereas miRNAs and antitargets are coex- number, with their genes distinguished by letter suffixes
pressed. Farh et al24 found similar results for targets and antitargets (eg, miR-181a).
of mammalian miRNAs, although in their analyses the predicted
targets were present at lower levels in tissues with the cognate
miRNAs and not excluded altogether. Furthermore, in a temporal miRNA biogenesis and mechanisms of
analysis of mouse myotube differentiation, they showed that posttranscriptional gene repression
predicted targets of tissue-specific miR-1 and miR-133 were highly
expressed before these miRNAs accumulated and subsequently, Mature miRNAs are 18 to 26 nucleotides in length, with 5⬘-
these targets were down-regulated. Together, these observations phosphate and 3⬘-hydroxyl groups. Their immediate precursors,
raise the possibility that some transcripts are predisposed to appear called pre-miRNAs, are characteristic stem-loop structures 60 to 80
or persist in domains where they are unnecessary or potentially bases in length with a 2-nucleotide 3⬘ overhang. In turn, pre-
harmful; miRNAs may have evolved to fine-tune gene expression miRNAs derive from long primary miRNA transcripts (pri-
programs and to eliminate gene products more rapidly than might miRNAs), which can exceed 1 kb and typically are products of
From www.bloodjournal.org by guest on March 11, 2018. For personal use only.

3648 SHIVDASANI BLOOD, 1 DECEMBER 2006 䡠 VOLUME 108, NUMBER 12

RNA polymerase II activity.29,30 Most pri-miRNAs are independent sors: long (hundreds to thousands of bases) double-stranded
transcriptional units, although many mammalian miRNAs are molecules in the case of siRNA and, in the case of miRNA, shorter
encoded within mRNA introns and appear to share the same (60- to 80-nucleotide) pre-miRNA stem-loop products. However,
primary transcripts as their protein-coding host genes. MicroRNAs both the production and functions of these diverse small RNAs
show highest evolutionary conservation in the stem, with variable require many common factors: endonucleases such as Dicer, which
divergence within the loop.26,31 Although conservation drops sharply in specifically cleave double-stranded RNA, and small RNA-binding
sequences flanking the miRNA hairpin,31 these regions are neverthe- proteins of the Argonaute family, which direct target molecules into
less required for nuclear processing of the primary transcript.15 the RISC ribonuclear complex.34 Moreover, the mRNA degrada-
Pri-miRNAs are first processed in the nucleus into 50- to tion apparatus, including RISC complexes, localizes in discrete
80-base pre-miRNA stem-loops by the Microprocessor complex of cytoplasmic “processing” or “P” bodies (also known as GW
nucleases and associated factors, including the RNase III Drosha bodies), where transcripts targeted by miRNAs were shown
and its partner DGCR8/Pasha (Figure 2).32,33 Pre-miRNA mol- recently to concentrate in a miRNA-dependent manner.43 Thus, like
ecules, in association with the Ran-GTP–dependent factor expor- siRNA-induced mRNA degradation, apparent repression of transla-
tin-5, are actively transported into the cytoplasm, where a second tion by miRNAs also involves delivery of transcripts to P-bodies,
RNase III–like nuclease, Dicer, processes them further into du- which lack ribosomal components; this relocation could be a
plexes that contain the 20- to 24-nucleotide mature miRNA and an consequence or, alternatively, serve as the basis for inhibiting
oligonucleotide of similar size from the other arm of the hair- protein translation.
pin.34-37 The nascent miRNA may be present on either arm of the
Early work suggested that siRNAs operate to eliminate their
stem-loop precursor, but each pre-miRNA yields a single mature
mRNA targets, whereas miRNAs commonly preserve target tran-
miRNA product. Gaps, bulges, and mismatches in the secondary
script levels and were thereby inferred to block protein translation.
structure of pre-miRNAs are predicted to reduce their stability and
The distinctions between siRNAs and miRNAs continue to blur,
to influence the efficiency by which they are processed to produce
and miRNAs are now recognized to facilitate both translational
mature miRNAs.38 The functional element in the precursor duplex
inhibition and mRNA degradation.44,45 Localization of the RISC
is determined when, after unwinding, one of the 2 strands is loaded
complex in P-bodies may help in understanding the apparent
asymmetrically into the RNA-induced silencing complex (RISC);
this tends to be the strand in which the 5⬘ end is less tightly paired differences, and miRNA-mediated transcript degradation may represent
to its complement.39,40 a secondary consequence of sequestration within P-bodies.3,43 Steady-
Similar thermodynamic considerations apply to the interactions state levels of transcripts with a high intrinsic decay rate may overtly
between mature miRNAs and their protein-coding RNA targets. reveal a substantial decline, whereas inherently stable mRNAs
Thus, the ability of a miRNA to repress cognate mRNAs is might persist in the cell; however, both transcript classes are
determined in large part by the free energy of binding of nucleo- probably equally subject to translational repression.
tides 2 to 8, counted from the 5⬘ end, with substantially lesser An insightful study recently shed light on several questions that
contribution from the 3⬘ region of the miRNA.41 Nucleotides 2 to 8 are pertinent to this review. After delivering miR-124 into HeLa
are hence regarded as the “seed” for miRNA target recognition, and cells, Lim and colleagues observed a shift in the expression profile
evolutionary conservation of complementarity with this relatively toward that of brain, the site where endogenous miR-124 is most
small region has served as a key basis for computational prediction abundant; similarly, transfection of miR-1 shifted the profile
of target genes. Conserved adenosine residues tend to flank the toward that of muscle, where miR-1 is preferentially expressed.45
complementary region, which implies that primary sequence determi- Taken at face value, even in the absence of a clear molecular
nants and base-pairing combine to specify target recognition.21,42 mechanism, these results implicate selected miRNAs in aspects of
In their size and chemistry, miRNAs resemble siRNAs, which gene regulation that are central to cellular identity. Notably, in each
in invertebrates recognize targets by precise complementarity and case, the levels of dozens of transcripts declined within 12 hours,
elicit their decay. As depicted in Figure 1, these 2 classes of small, and the 3⬘-UTRs of affected mRNAs were enriched for sequences
noncoding RNAs derive from distinct classes of immediate precur- complementary to the 5⬘ ends of the cognate miRNAs. Thus, some

Figure 2. miRNA biogenesis and action. Most pri-mRNAs are the


products of independent genes, transcribed by RNA polymerase II.
The nuclear Microprocessor protein complex, which contains the
RNase III Drosha and its partner DGCR8/Pasha, cleaves pri-miRNAs
into 50- to 80-base pre-miRNA stem-loop moieties. The Ran-GTP–
dependent factor exportin-5 actively transports pre-miRNAs into the
cytoplasm, where the nuclease Dicer processes them further into
duplexes that contain the 20- to 24-nucleotide mature miRNA; each
pre-miRNA usually yields a single mature miRNA product. The
functional strand is determined when one of the 2 strands of the duplex
is loaded into the RISC, which contains Argonaute and related
proteins and localizes in cytoplasmic P-bodies. Recognition of target
mRNAs by partial sequence complementarity to the miRNA results in
posttranscriptional gene repression by some combination of transcript
degradation and translational inhibition.
From www.bloodjournal.org by guest on March 11, 2018. For personal use only.

BLOOD, 1 DECEMBER 2006 䡠 VOLUME 108, NUMBER 12 miRNA REGULATION OF HEMATOPOIESIS 3649

of the down-regulated transcripts may be targets for direct miRNA- prediction of hundreds of miRNAs.52,53 In an especially thorough
mediated degradation, an effect distinct from the purportedly main approach that integrated bioinformatic prediction, microarray anal-
role of miRNAs in inhibiting protein translation. Moreover, at least ysis, and sequence-directed cloning, Bentwich and coworkers
miR-1 and miR-124, but probably other tissue-specific miRNAs, identified nearly 100 new miRNAs and predicted the presence of at
seem to regulate a large number of transcripts directly. It is thus least 800 miRNAs in aggregate.54 However, the scope of potential
conceivable that establishing or maintaining cell identity depends miRNA regulation remains uncertain, and predicted miRNAs should be
in part on fine regulation of lineage-specific gene expression by a regarded cautiously until they are confirmed by experiments.
few key miRNAs. To add further complexity, miRNAs share limited sequence
Cardinal differentiation functions have traditionally been attrib- complementarity with their mRNA targets, so that each miRNA
uted to lineage-restricted transcription factors (TFs); either the TFs could possibly interact with scores of genes; conversely, a single
themselves or their tissue-specific outputs, or both, are hence the
gene can harbor multiple miRNA recognition sites within 1 or more
best candidates for direct miRNA regulation. In this light, it is
potential 3⬘-UTRs. Published studies in vertebrate species center
interesting that although current predictions of putative miRNA
on the results of bioinformatic predictions, and no mammalian
targets cover a broad range of molecular functions,20,21 the early
miRNA target has yet been identified prospectively without the
sets are enriched for TF-coding genes,42,46 and validated examples
benefit of computationally derived clues. The algorithms used to
of miRNA function also emphasize their roles in regulating
tissue-restricted TFs such as NF-I, Hand2, and HoxB8.18,19,44,47 predict miRNA targets typically develop scoring schemes based on
However, these examples reflect a certain investigation bias and sequence complementarity, free energy calculations of RNA duplex
miRNA functions clearly extend beyond targeting of TF tran- formation, and phylogenetic conservation. In a 4-genome analysis
scripts, as would be expected if miRNAs as a class can affect of 3⬘-UTRs, Lewis et al used the TargetScanS algorithm to identify
hundreds or thousands of genes.23,24 A well-studied counterex- 13 000 putative regulatory relationships conserved among the 4
ample is offered by the evolutionarily conserved, pancreatic islet species, thereby implicating more than 5 000 human genes (30% of
cell–specific miR-375, which seems to inhibit glucose-induced the tested gene set) as candidate miRNA targets.42 Using the
insulin secretion by targeting the myotrophin message and a independent PicTar algorithm, Krek et al reached similar conclu-
resultant effect on insulin exocytosis.48 sions on the scope of conserved miRNA targeting in mammals,
Independent of the possibly widespread role of miRNAs in with a set of predictions that largely overlaps that of the Tar-
suppressing TF genes, functional parallels between TFs and getScanS analysis.20 By contrast, John et al have predicted about
miRNAs as molecular regulators are noteworthy, as articulated in a 2 000 human gene targets, which currently have little overlap with
recent review.49 Both classes of molecules exert their activity those of TargetScanS and PicTar.46 Lewis et al detected additional
through composite cis-regulatory elements and function in combi- targeting in open reading frames and concluded that more than
nations; single TFs or miRNAs may regulate many different genes, one-third of human genes are under selective pressure to maintain
as we encountered for miR-1 and miR-124. TFs and miRNAs also pairing to miRNAs.42 Furthermore, there is increasing experimen-
may act cooperatively on their target genes, which typically harbor tal and computational evidence for a large number of nonconserved
multiple cis-regulatory DNA elements for transcriptional activation targets.23,24,45 However, individual predictions of miRNA targets
and more than 1 putative miRNA recognition site for mRNA should be regarded as provisional, and attempts at biological
down-regulation.20,21,50 Thus, both TFs and miRNAs may generate investigation should be preceded by careful experimental valida-
combinatorial codes that dictate cellular identities and phenotypes.
tion of putative miRNA-target combinations.
Understanding the putative networks of these interactions is a
The possibility that a single miRNA may target multiple
central challenge in studying miRNA functions in cell differentiation.
transcripts and that individual transcripts may be subject to
regulation by multiple miRNAs amplifies the scope of putative
miRNA regulation in potentially unfathomable terms. The concep-
How many miRNAs? How many targets? tual and bioinformatic problems presented by these considerations
are formidable and may be compared with those that follow from
To appreciate the breadth of miRNA functions, it would be useful genome-scale identification of protein-protein interactions. In the
to have reliable estimates of the total number of miRNA genes in a latter example, however, mapping the domains of gene expression
species. After the initial cloning of more than 100 miRNAs,7-10 reduces the magnitude of the challenge as proteins can legitimately
cloning efforts soon achieved a degree of saturation, with recurring
interact only when they are coexpressed. In contrast, miRNAs may
identification of previously cloned species. However, miRNAs that
be coexpressed with their targets to dampen gene expression, or the
are expressed at low abundance, restricted to a few cells in an
2 may appear in complementary distributions when the miRNA
organ, developmentally regulated, or technically resistant to clon-
serves to eliminate target gene expression. Resolution of the
ing might elude detection in the common strategies. Likewise, even
the most sensitive algorithms to predict miRNA genes make underlying questions will require rigorous experimentation and
assumptions in order to enforce stringency in computational confident assessment of low levels of miRNA and mRNA products.
searches. Nevertheless, a combination of comprehensive cloning In addition, it is likely that not all miRNAs function similarly: some
and computer prediction has expanded the number of animal may indeed regulate a single gene tightly, whereas others may have
miRNAs greatly. Most cloned miRNAs show sequence conserva- a wider influence over gene expression programs associated with
tion among closely related species and also across large evolution- selected developmental or physiologic states (Figure 3).
ary distances: among the initially identified mouse and human Before reviewing what little is known about miRNA influences
miRNAs, more than half have identifiable homologs in fish, and in hematopoiesis, it is worthwhile to explore the general lessons we
about half of these have homologs in flies or worms.51 It is this might learn from the few reported examples of mammalian miRNA
evolutionary conservation, coupled with consideration of stem- functions. Mouse miR-196, which is encoded at 3 paralogous
loop structures, that provides an important basis for computational locations in the mammalian A, B, and C Hox gene clusters,
From www.bloodjournal.org by guest on March 11, 2018. For personal use only.

3650 SHIVDASANI BLOOD, 1 DECEMBER 2006 䡠 VOLUME 108, NUMBER 12

Figure 3. Models for the biological significance of


miRNA functions. Three nonmutually exclusive models
are presented, and the truth is likely to incorporate
elements from each. Different modes may also operate in
different cells or at different stages in development.
Computational predictions form the basis for much of the
current thinking and experimental evidence favoring each
of the models remains limited.

represses HoxB8 mRNA and downstream Sonic hedgehog signal- a limited number of other sites. miR-223 and miR-142 showed the
ing, factors that distinguish developing forelimbs from hind- most restricted distribution, virtually confined to bone marrow and
limbs.18,44 In a different scenario, the miR-1 genes are direct spleen; some miR-142 expression is also seen in thymus, where
transcriptional targets of muscle differentiation TFs such as serum miR-181a is especially abundant.15 Other results from miRNA
response factor, MyoD, and Mef2, and their miRNA products microarray and Northern analysis of assorted blood cell (mainly
appear to regulate levels of Hand2, a TF that promotes cardiomyo- lymphocyte) populations suggest that few miRNAs are truly
cyte expansion in the cardiac ventricles; overexpression of miR-1 restricted in distribution.55 Indeed, tissue specificities reported for
in the developing heart reduces the pool size of proliferating miRNA expression in general probably reflect enrichment, which
ventricular cardiomyocytes.19 In addition to a third instance that is can even be considerable, in certain cells and not necessarily
described in the next section, “miRNA expression and functions in exclusion from all other sites. Nevertheless, some miRNAs are
hematopoiesis,”47 these examples share certain common themes remarkably responsive to cellular manipulations: miR-150 is
that may be instructive. First, in each case, the miRNA under rapidly down-regulated upon T-cell stimulation under T helper 1
detailed investigation shows significant differential expression in a (Th1) or Th2 conditions, whereas miR-146 is selectively increased
manner that permitted generation of cogent and testable hypoth- in Th1 lymphocytes.55 In a recent expression analysis of megakaryo-
eses. miR-1 is highly enriched in cardiac and skeletal muscle cyte cultures derived from CD34⫹ bone marrow cells, Garzon and
precursors, whereas miR-196 expression is at least 20-fold higher colleagues identified 19 miRNAs that were reduced by 2- to
in hindlimb buds than in the forelimbs. A second, related point is 50-fold and none that increased with megakaryocytic differentia-
that in each case there was already considerable information about tion.56 Although 2 of these miRNAs, miR-10a and miR-130a, are
the tissue under study so that the investigators could test for thought to target the transcription factor genes HOXA1 and MAFB,
consistency and rely on experimental systems that are well respectively,56 the true significance of most miRNA differential
validated. Third, identification of miRNA targets remains heavily expression or of changes induced in hematopoiesis is not known.
reliant on computational prediction, again coupled with prior To interrogate miRNA functions in hematopoiesis, Chen et al15
knowledge that helped focus on plausible candidates. These themes introduced the blood cell–specific miRNAs they identified into
will likely continue to guide further work in biological miRNA murine Lin⫺ hematopoietic progenitor cells, followed by in vitro
functions into the foreseeable future. Of course, these examples differentiation or transplantation into lethally irradiated hosts.
illustrate the seminal role of selected factors that may have a Overexpression of miR-181a induced a 2-fold increase in B
limited repertoire of TF targets. This scenario is quite possibly the lymphocyte numbers without affecting the T-cell lineage in vitro,
exception rather than the rule for general miRNA functions. It will although the mouse transplantation studies yielded a nearly 2-fold
require new experimental paradigms to gain better insights into reduction in the number of circulating T lymphocytes and about a
cases where a few or many miRNAs operate in combination with 90% decrease in the CD8⫹ subpopulation. In contrast, miR-142s
innumerable targets to fine-regulate biological processes. and miR-223, both evaluated only in vitro, modestly increased
T-cell and not B-cell numbers; miR-30, which is not preferentially
expressed in blood cells, did not affect hematopoiesis as judged by
miRNA expression and functions flow cytometry for lineage markers. Whereas miR-181a hence
in hematopoiesis seems capable of modulating lymphoid cell lineages, it is important
to note that these conclusions rely exclusively on studies of
Hematopoietic cells were not included in the earliest reported overexpressed miRNAs. Indeed, experimental constraints limit the
efforts to identify tissue-restricted miRNAs.10 Subsequently, Chen ability to assess miRNA functions rapidly through loss of function,
and colleagues15 cloned about 100 unique miRNAs from mouse and in any event, it is likely that regulation by miRNAs encom-
bone marrow. Whereas many had been identified previously from passes considerable redundancy.
nonhematopoietic tissues, a select few, including miR-142, miR- In contrast to the specific requirement of selected miRNAs, the
181a, and miR-223, are expressed preferentially in blood cells and general requirement for miRNAs in a hematopoietic organ has been
From www.bloodjournal.org by guest on March 11, 2018. For personal use only.

BLOOD, 1 DECEMBER 2006 䡠 VOLUME 108, NUMBER 12 miRNA REGULATION OF HEMATOPOIESIS 3651

evaluated in mice with selective deletion of the dcr-1 (Dicer) gene moter, and as long as NFI-A is present, this miR gene is inactive.
in the thymus.57 These mice displayed a severe block in develop- With the appearance of C/EBP␣, which occurs upon retinoic
ment of peripheral CD8⫹ T cells and reduced numbers of CD4⫹ T acid–induced commitment to granulocytic differentiation, miR-223
lymphocytes, which also proliferated poorly or died by apoptosis expression is activated, resulting in repression of its NFI-A mRNA
quickly after agonist stimulation. Dicer-deficient CD4⫹ T cells target. Thus, miR-223 appears to be a key player in a simple
preferentially expressed interferon-␥, a cytokine affiliated with the regulatory circuit of TFs that control granulopoiesis and may help
Th1 lineage. These observations represent the net effect of miRNA stabilize the neutrophil phenotype induced by C/EBP␣ (Figure 4).
deficiency and perhaps other unknown Dicer functions, possibly As NFI-A mRNA is just one among literally hundreds of possible
including processing of siRNA. Accordingly, it remains unclear miR-223 targets, the particular circuit described here may represent
whether they reflect tissue-specific absence of a single critical only a facet of its full molecular effects. In principle, miR-223
miRNA, of a few regulators that control a limited number of could serve to down-regulate many genes that would ordinarily be
mRNA targets, or the functional output of a complex pathway expressed in multipotential progenitors or nongranulocytic cells
consisting of many miRNAs, each with a defined individual role. and thus help refine the neutrophil-specific gene expression program.
Indeed, the fundamental limitation of both the miR-181 overexpres- Although this hypothesis remains to be tested, it is consistent both
sion and Dicer gene deletion studies is that the identity and with emerging concepts in the field and with the demonstration that
activities of the primary mRNA targets are unknown. Target particular miRNAs can influence discrete aspects of hematopoiesis.
prediction algorithms identify no fewer than 340 conserved candi- In a similar vein, Felli and colleagues62 found high expression
date targets for miR-181a, with equivalent degrees of confidence of miR-221 and miR-222 in human cord blood–derived CD34⫹
for dozens of them. Thus, dissecting the regulatory circuit poses an hematopoietic progenitor cells; unilineage erythroid differentiation
enormous challenge. By contrast, the miRanda algorithm,46 whose of these cells in culture caused reduction in the levels of these 2
predicted targets overlap minimally with others’, hints at the miRNAs, which have virtually identical sequence in the seed
human HOXA5 gene as a candidate miR-181 target. This putative region of target pairing. Bioinformatic analysis suggested c-kit as a
target gene is expressed in hematopoietic cells, where it has possible target transcript, and c-kit mRNA levels correlate in-
previously been implicated in regulating the balance between versely with those of miR-221 and miR-222 in cultured cells.
myeloid and erythroid cell precursors.58,59 The latter studies did Forced expression of these miRNAs in TF-1 erythroleukemia cells
not, however, report lymphocyte ratios; miR-181a overexpression or in erythroid culture of CD34⫹ cord blood cells slowed cell
does not influence erythroid cell differentiation;15 and interaction growth concomitant with reduced c-kit protein levels. Although
between miR-181a and HOXA5 remains speculative at best. these results do not address the true functions of the X-linked
miR-223 is another blood cell–restricted miRNA, which achieves miR-221/miR-222 cluster in vivo, the authors’ approach and the
highest levels in CD34⫺ bone marrow cells, is not present in T or B internal consistency of the findings highlight current experimental
lymphocytes, and is induced by retinoic acid treatment of myeloid paradigms to link specific, tissue-restricted miRNAs with candi-
precursors through the transcriptional activator C/EBP␣.47 C/EBP␣ date mRNA targets. In the future, such studies will need to be
is known to be important in myelopoiesis as it regulates many extended in 2 complementary ways: targeted disruption of indi-
myeloid cell–specific genes and promotes granulocyte-specific vidual miRNA genes and targeted mutation of putative miRNA-
genes at the expense of the monocytic differentiation program in binding sites in the 3⬘-UTRs of candidate targets. While redun-
bipotential precursors60; C/EBP␣ knockout mice show selective dancy among miRNA regulators may limit the force of such
failure of neutrophil and eosinophil differentiation.61 Overexpres- experiments, a thorough understanding of miRNA functions will
sion of miR-223 in the undifferentiated NB4 cell line induced only emerge from integrated consideration of complementary
morphologic changes consistent with maturation and expression of results. miRNAs implicated to date in specific hematopoietic
CD11b and the granulocyte colony-stimulating factor receptor. functions are summarized in Table 1.
Conversely, in one of the few examples where a vertebrate miRNA
function has been evaluated through experimental reduction in its
levels, miR-223–complementary locked nucleic acid oligonucleo- miRNA expression and functions in leukemia
tides induced modest reductions in the amounts of both miR-223
and CD11b.47 These miR-223 effects are mediated in part through Naturally, there has been considerable interest in any connection
translational inhibition of mRNA for the TF NFI-A. In a particu- between miRNAs and cancer, including leukemias. The miR-15a/
larly provocative twist, NFI-A and C/EBP␣ compete for binding to miR-16 locus lies within a region of human chromosome 13 that is
partially overlapping DNA sequences in the miR-223 gene pro- also the site of the most common structural abnormality in B-cell

Figure 4. Working model for the regulatory circuit involving


miR-223, C/EBP␣, and NFI-A in neutrophil differentiation. Experi-
mental results47 are consistent with the idea that NFI-A and C/EBP␣
compete for binding to partially overlapping DNA sequences in the
miR-223 gene promoter, and as long as NFI-A is present, this miR
gene is inactive. Cells committed to granulocytic differentiation ex-
press C/EBP␣, which activates miR-223 expression, resulting in
repression of its NFI-A mRNA target. Thus, miR-223 appears to
participate in a simple regulatory circuit of TFs that control granulopoi-
esis and may help stabilize the neutrophil phenotype induced by
C/EBP␣. Reprinted from Fazi et al47 with permission.
From www.bloodjournal.org by guest on March 11, 2018. For personal use only.

3652 SHIVDASANI BLOOD, 1 DECEMBER 2006 䡠 VOLUME 108, NUMBER 12

Table 1. miRNAs implicated to date in hematopoietic functions


miRNA Candidate target genes Function Source

miR-181a None identified Lymphopoiesis Chen et al15


miR-223 NFI-A Granulopoiesis Fazi et al47
miR-221, miR-222 c-kit Erythropoiesis Felli et al62
miR-130a MafB Megakaryopoiesis (assumed) Garzon et al56
miR-10a HoxA1 Megakaryopoiesis (assumed) Garzon et al56

chronic lymphocytic leukemia (CLL) and mantle cell lymphoma; they regulate broad programs of unique gene expression through
both miRNAs are deleted or down-regulated in nearly two-thirds of minor effects on multiple targets, as suggested by evolutionary
CLL samples.63 Other miRNA genes also are frequently located at considerations and some experimental results? What is the larger
fragile sites, minimal regions of loss of heterozygosity or amplifica- role of miRNAs in human disease and do they provide novel
tion, and common breakpoint regions previously identified in a therapeutic opportunities? As these questions begin to capture
range of human malignancies.64 Calin and colleagues65 identified investigators’ attention, it is worth recalling the seminal contribu-
germline or somatic mutations in 5 of 42 sequenced miRNAs in 11 tions of hematologists and their particular models in solving
of 75 patients with CLL and no such mutations in 160 subjects problems in genetics, cell differentiation, and biochemistry. Inas-
without cancer. Whereas these observations provoke speculation much as miRNAs may control molecular aspects of cell specializa-
about direct miRNA functions in leukemogenesis, other advances tion, hematopoiesis offers an ideal model system in which ideas
center on more general features of miRNA expression in the about their mechanisms and functions can be tested incisively.
disease context. The mixed-lineage leukemia (MLL) 1 transcrip- Meanwhile, the apparent scope of miRNA regulation has disturbed
tional regulator binds to transcription-start sites of innumerable simple and conventional views about the classes of candidate
genes, including 30% of all tested miRNA loci,66 which suggests regulators in hematopoiesis and hematologic disorders. No doubt
that broad dysregulation of both mRNA and miRNA expression the field will rapidly embrace the new biology and endeavor to
may underlie leukemic phenotypes. Lu and coworkers67 report understand how and to what degree miRNAs influence different
widespread down-regulation of miRNAs in tumors compared with aspects of blood cell renewal and specialization. By the time these
their normal tissue counterparts, much as others found reduced problems are solved, we can look forward to other discoveries that
levels of specific miRNAs in colon cancer specimens.68 In one might inform us about the functions of the vast majority of the
study, miRNA expression profiles were more successful than genome that encodes neither proteins nor other known regulatory
mRNA expression in classifying poorly differentiated tumors.67 molecules like miRNAs.
Furthermore, a unique expression profile consisting of 13 miRNA
genes could distinguish cases of CLL with distinct molecular
markers and clinical outcomes.65 The broader role of miRNAs in Acknowledgments
classifying tumors or contributing to disease progression remains
to be defined and is likely to be an area of intense investigation. I am grateful to Jochen Hartner, David Nathan, and Stuart Orkin for
critical review of the manuscript and to Alice Chen for help in
preparing figures. As Blood guidelines limit the number of cited
Concluding remarks references, I regret the highly selective (and sometimes arbitrary)
citation process I needed to adopt in writing a succinct review.
Progress in the area of miRNAs seems to have occurred faster than This work was supported in part by grants from the National
in any major biological discipline in recent memory. The field has Institutes of Health.
moved from virtual ignorance about an abundant class of regula-
tory molecules to a reasonably advanced understanding of the
mechanisms of miRNA biogenesis and an emerging consensus Authorship
about the numbers of miRNAs and their targets in several species,
including humans. The next series of challenges is already becom- The author declares no competing financial interests.
ing evident: how is transcription of miRNA genes controlled? Do Correspondence: Ramesh A. Shivdasani, Dana-Farber Cancer
miRNAs operate more commonly through single, critical target Institute, 44 Binney St, Boston, MA 02115; e-mail:
transcripts, as the examples reported to date would imply, or do ramesh_shivdasani@dfci.harvard.edu.

References
1. Bartel DP. MicroRNAs: genomics, biogenesis, nucleotide let-7 RNA regulates developmental 10. Lagos-Quintana M, Rauhut R, Yalcin A, Meyer J,
mechanism, and function. Cell. 2004;116:281-297. timing in Caenorhabditis elegans. Nature. 2000; Lendeckel W, Tuschl T. Identification of tissue-
2. Wienholds E, Plasterk RH. MicroRNA function in 403:901-906. specific microRNAs from mouse. Curr Biol. 2002;
animal development. FEBS Lett. 2005;579:5911- 7. Lagos-Quintana M, Rauhut R, Lendeckel W, Tus- 12:735-739.
5922. chl T. Identification of novel genes coding for 11. Ambros V, Bartel B, Bartel DP, et al. A uniform
3. Zamore PD, Haley B. Ribo-gnome: the big world small expressed RNAs. Science. 2001;294:853- system for microRNA annotation. RNA. 2003;9:
of small RNAs. Science. 2005;309:1519-1524. 858. 277-279.
4. Ambros V. The functions of animal microRNAs. 8. Lau NC, Lim LP, Weinstein EG, Bartel DP. An 12. Chen PY, Manninga H, Slanchev K, et al. The de-
Nature. 2004;431:350-355. abundant class of tiny RNAs with probable regu- velopmental miRNA profiles of zebrafish as deter-
5. Lee RC, Feinbaum RL, Ambros V. The C. elegans latory roles in Caenorhabditis elegans. Science. mined by small RNA cloning. Genes Dev. 2005;
heterochronic gene lin-4 encodes small RNAs 2001;294:858-862. 19:1288-1293.
with antisense complementarity to lin-14. Cell. 9. Lee RC, Ambros V. An extensive class of small 13. Giraldez AJ, Cinalli RM, Glasner ME, et al. Mi-
1993;75:843-854. RNAs in Caenorhabditis elegans. Science. 2001; croRNAs regulate brain morphogenesis in ze-
6. Reinhart BJ, Slack FJ, Basson M, et al. The 21- 294:862-864. brafish. Science. 2005;308:833-838.
From www.bloodjournal.org by guest on March 11, 2018. For personal use only.

BLOOD, 1 DECEMBER 2006 䡠 VOLUME 108, NUMBER 12 miRNA REGULATION OF HEMATOPOIESIS 3653

14. Wienholds E, Kloosterman WP, Miska E, et al. non GJ. Processing of primary microRNAs by the Computational identification of Drosophila mi-
MicroRNA expression in zebrafish embryonic de- Microprocessor complex. Nature. 2004;432:231- croRNA genes. Genome Biol. 2003;4:R42.
velopment. Science. 2005;309:310-311. 235. 53. Lim LP, Glasner ME, Yekta S, Burge CB, Bartel
15. Chen CZ, Li L, Lodish HF, Bartel DP. MicroRNAs 34. Grishok A, Pasquinelli AE, Conte D, et al. Genes DP. Vertebrate microRNA genes. Science. 2003;
modulate hematopoietic lineage differentiation. and mechanisms related to RNA interference 299:1540.
Science. 2004;303:83-86. regulate expression of the small temporal RNAs 54. Bentwich I, Avniel A, Karov Y, et al. Identification
16. Liu CG, Calin GA, Meloon B, et al. An oligonucle- that control C. elegans developmental timing. of hundreds of conserved and nonconserved hu-
otide microchip for genome-wide microRNA profil- Cell. 2001;106:23-34. man microRNAs. Nat Genet. 2005;37:766-770.
ing in human and mouse tissues. Proc Natl Acad 35. Hutvagner G, McLachlan J, Pasquinelli AE, Balint 55. Monticelli S, Ansel KM, Xiao C, et al. MicroRNA
Sci U S A. 2004;101:9740-9744. E, Tuschl T, Zamore PD. A cellular function for the profiling of the murine hematopoietic system. Ge-
17. Wienholds E, Koudijs MJ, van Eeden FJ, Cuppen RNA-interference enzyme Dicer in the maturation nome Biol. 2005;6:R71.
E, Plasterk RH. The microRNA-producing en- of the let-7 small temporal RNA. Science. 2001;
56. Garzon R, Pichiorri F, Palumbo T, et al. MicroRNA
zyme Dicer1 is essential for zebrafish develop- 293:834-838.
fingerprints during human megakaryocytopoiesis.
ment. Nat Genet. 2003;35:217-218. 36. Lund E, Guttinger S, Calado A, Dahlberg JE, Proc Natl Acad Sci U S A. 2006;103:5078-5083.
18. Hornstein E, Mansfield JH, Yekta S, et al. The Kutay U. Nuclear export of microRNA precursors.
57. Muljo SA, Ansel KM, Kanellopoulou C, Livingston
microRNA miR-196 acts upstream of Hoxb8 and Science. 2004;303:95-98.
DM, Rao A, Rajewsky K. Aberrant T cell differen-
Shh in limb development. Nature. 2005;438:671- 37. Yi R, Qin Y, Macara IG, Cullen BR. Exportin-5 tiation in the absence of Dicer. J Exp Med. 2005;
674. mediates the nuclear export of pre-microRNAs 202:261-269.
19. Zhao Y, Samal E, Srivastava D. Serum response and short hairpin RNAs. Genes Dev. 2003;17:
58. Crooks GM, Fuller J, Petersen D, et al. Constitu-
factor regulates a muscle-specific microRNA that 3011-3016.
tive HOXA5 expression inhibits erythropoiesis
targets Hand2 during cardiogenesis. Nature. 38. Hutvagner G, Zamore PD. A microRNA in a mul- and increases myelopoiesis from human hemato-
2005;436:214-220. tiple-turnover RNAi enzyme complex. Science. poietic progenitors. Blood. 1999;94:519-528.
20. Krek A, Grun D, Poy MN, et al. Combinatorial mi- 2002;297:2056-2060.
59. Fuller JF, McAdara J, Yaron Y, Sakaguchi M,
croRNA target predictions. Nat Genet. 2005;37: 39. Khvorova A, Reynolds A, Jayasena SD. Func- Fraser JK, Gasson JC. Characterization of HOX
495-500. tional siRNAs and miRNAs exhibit strand bias. gene expression during myelopoiesis: role of
21. Lewis BP, Burge CB, Bartel DP. Conserved seed Cell. 2003;115:209-216. HOX A5 in lineage commitment and maturation.
pairing, often flanked by adenosines, indicates 40. Schwarz DS, Hutvagner G, Du T, Xu Z, Aronin N, Blood. 1999;93:3391-3400.
that thousands of human genes are microRNA Zamore PD. Asymmetry in the assembly of the
targets. Cell. 2005;120:15-20. 60. Radomska HS, Huettner CS, Zhang P, Cheng T,
RNAi enzyme complex. Cell. 2003;115:199-208. Scadden DT, Tenen DG. CCAAT/enhancer bind-
22. Bartel DP, Chen CZ. Micromanagers of gene ex- ing protein alpha is a regulatory switch sufficient
41. Doench JG, Sharp PA. Specificity of microRNA
pression: the potentially widespread influence of for induction of granulocytic development from
target selection in translational repression. Genes
metazoan microRNAs. Nat Rev Genet. 2004;5: bipotential myeloid progenitors. Mol Cell Biol.
Dev. 2004;18:504-511.
396-400. 1998;18:4301-4314.
42. Lewis BP, Shih IH, Jones-Rhoades MW, Bartel
23. Stark A, Brennecke J, Bushati N, Russell RB, Co-
DP, Burge CB. Prediction of mammalian mi- 61. Zhang DE, Zhang P, Wang ND, Hetherington CJ,
hen SM. Animal microRNAs confer robustness to
croRNA targets. Cell. 2003;115:787-798. Darlington GJ, Tenen DG. Absence of granulo-
gene expression and have a significant impact on
43. Liu J, Valencia-Sanchez MA, Hannon GJ, Parker cyte colony-stimulating factor signaling and neu-
3⬘ UTR evolution. Cell. 2005;123:1133-1146.
R. MicroRNA-dependent localization of targeted trophil development in CCAAT enhancer binding
24. Farh KK, Grimson A, Jan C, et al. The wide- protein alpha-deficient mice. Proc Natl Acad Sci
mRNAs to mammalian P-bodies. Nat Cell Biol.
spread impact of mammalian MicroRNAs on U S A. 1997;94:569-574.
2005;7:719-723.
mRNA repression and evolution. Science. 2005;
44. Yekta S, Shih IH, Bartel DP. MicroRNA-directed 62. Felli N, Fontana L, Pelosi E, et al. MicroRNAs
310:1817-1821.
cleavage of HOXB8 mRNA. Science. 2004;304: 221 and 222 inhibit normal erythropoiesis and
25. Hu M, Krause D, Greaves M, et al. Multilineage erythroleukemic cell growth via kit receptor down-
594-596.
gene expression precedes commitment in the modulation. Proc Natl Acad Sci U S A. 2005;102:
hemopoietic system. Genes Dev. 1997;11:774- 45. Lim LP, Lau NC, Garrett-Engele P, et al. Microar- 18081-18086.
785. ray analysis shows that some microRNAs down-
regulate large numbers of target mRNAs. Nature. 63. Calin GA, Dumitru CD, Shimizu M, et al. Frequent
26. Lim LP, Lau NC, Weinstein EG, et al. The micro- deletions and down-regulation of micro-RNA
RNAs of Caenorhabditis elegans. Genes Dev. 2005;433:769-773.
genes miR15 and miR16 at 13q14 in chronic lym-
2003;17:991-1008. 46. John B, Enright AJ, Aravin A, Tuschl T, Sander C, phocytic leukemia. Proc Natl Acad Sci U S A.
27. Wellcome Trust Genome Campus. miRBase. Marks DS. Human MicroRNA targets. PLoS Biol. 2002;99:15524-15529.
http://microrna.sanger.ac.uk/. Accessed May 30, 2004;2:e363.
64. Calin GA, Sevignani C, Dumitru CD, et al. Human
2006. 47. Fazi F, Rosa A, Fatica A, et al. A minicircuitry microRNA genes are frequently located at fragile
28. Griffiths-Jones S. The microRNA registry. Nucleic comprised of microRNA-223 and transcription sites and genomic regions involved in cancers.
Acids Res. 2004;32:D109-D111. factors NFI-A and C/EBPalpha regulates human Proc Natl Acad Sci U S A. 2004;101:2999-3004.
granulopoiesis. Cell. 2005;123:819-831.
29. Lee Y, Kim M, Han J, et al. MicroRNA genes are 65. Calin GA, Ferracin M, Cimmino A, et al. A Mi-
transcribed by RNA polymerase II. EMBO J. 48. Poy MN, Eliasson L, Krutzfeldt J, et al. A pancre- croRNA signature associated with prognosis and
2004;23:4051-4060. atic islet-specific microRNA regulates insulin se- progression in chronic lymphocytic leukemia.
cretion. Nature. 2004;432:226-230. N Engl J Med. 2005;353:1793-1801.
30. Cai X, Hagedorn CH, Cullen BR. Human micro-
RNAs are processed from capped, polyadenyl- 49. Hobert O. Common logic of transcription factor 66. Guenther MG, Jenner RG, Chevalier B, et al.
ated transcripts that can also function as mRNAs. and microRNA action. Trends Biochem Sci. 2004; Global and Hox-specific roles for the MLL1 meth-
RNA. 2004;10:1957-1966. 29:462-468. yltransferase. Proc Natl Acad Sci U S A. 2005;
31. Berezikov E, Guryev V, van de Belt J, Wienholds 50. Lai EC, Tam B, Rubin GM. Pervasive regulation 102:8603-8608.
E, Plasterk RH, Cuppen E. Phylogenetic shadow- of Drosophila Notch target genes by GY-box-, 67. Lu J, Getz G, Miska EA, et al. MicroRNA expres-
ing and computational identification of human mi- Brd-box-, and K-box-class microRNAs. Genes sion profiles classify human cancers. Nature.
croRNA genes. Cell. 2005;120:21-24. Dev. 2005;19:1067-1080. 2005;435:834-838.
32. Lee Y, Ahn C, Han J, et al. The nuclear RNase III 51. Lagos-Quintana M, Rauhut R, Meyer J, Borkhardt 68. Michael MZ, O’Connor SM, van Holst Pellekaan
Drosha initiates microRNA processing. Nature. A, Tuschl T. New microRNAs from mouse and NG, Young GP, James RJ. Reduced accumula-
2003;425:415-419. human. RNA. 2003;9:175-179. tion of specific microRNAs in colorectal neopla-
33. Denli AM, Tops BB, Plasterk RH, Ketting RF, Han- 52. Lai EC, Tomancak P, Williams RW, Rubin GM. sia. Mol Cancer Res. 2003;1:882-891.
From www.bloodjournal.org by guest on March 11, 2018. For personal use only.

2006 108: 3646-3653


doi:10.1182/blood-2006-01-030015 originally published
online August 1, 2006

MicroRNAs: regulators of gene expression and cell differentiation


Ramesh A. Shivdasani

Updated information and services can be found at:


http://www.bloodjournal.org/content/108/12/3646.full.html
Articles on similar topics can be found in the following Blood collections
Free Research Articles (4923 articles)
Gene Expression (1086 articles)
Hematopoiesis and Stem Cells (3486 articles)
Immunobiology and Immunotherapy (5568 articles)
Neoplasia (4182 articles)
Reviews in Translational Hematology (58 articles)
Signal Transduction (1930 articles)
Stem Cells in Hematology (166 articles)

Information about reproducing this article in parts or in its entirety may be found online at:
http://www.bloodjournal.org/site/misc/rights.xhtml#repub_requests

Information about ordering reprints may be found online at:


http://www.bloodjournal.org/site/misc/rights.xhtml#reprints

Information about subscriptions and ASH membership may be found online at:
http://www.bloodjournal.org/site/subscriptions/index.xhtml

Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by the American Society
of Hematology, 2021 L St, NW, Suite 900, Washington DC 20036.
Copyright 2011 by The American Society of Hematology; all rights reserved.

You might also like