You are on page 1of 10

REVIEWS

CNS pRECuRSoRS

Roles of small regulatory RNAs


in determining neuronal identity
Xuekun Li and Peng Jin
Abstract | Neurogenesis, the process of generating functional neurons from neural stem cells,
is tightly controlled by many intrinsic and extrinsic mechanisms. Uncovering these regulatory
mechanisms is crucial for understanding the functions and plasticity of the human brain.
Recent studies in both invertebrates and vertebrates point to the importance of small
regulatory RNAs in regulating lineage-specific gene expression and determining neuronal
identity during neurogenesis. These new observations suggest that small regulatory RNAs
could function at many levels to regulate self-renewal of neural stem cells and neuronal fate
specification, implicating small regulatory RNAs in the complexity of neurogenesis.

Neurogenesis is the process by which functionally levels and through a range of mechanisms. Uncovering
Self-renewal
The capacity of a cell to integrated neurons are generated from neural stem cells the functions of these non-coding RNAs could substan-
proliferate and produce (NSCs). It involves the proliferation and neuronal fate tially improve our understanding and the treatment of
identical cells. specification of NSCs, as well as the maturation and human diseases.
functional integration of the neuronal progeny into neu- The small regulatory RNAs identified so far include
Multipotency
The potential of a cell to give
ronal circuits1,2. NSCs exhibit the two essential proper- microRNAs (miRNAs), endogenous small interfering
rise to multiple lineage cells. ties of stem cells: self-renewal and multipotency. During RNAs (esiRNAs), Piwi-interacting RNAs (piRNAs) and
Neural stem cells, for example embryogenesis, NSCs are located in the ventricular zone promoter-associated short RNAs (PASRs; also known as
can generate neurons, of the neural tube. Embryonic NSCs can give rise to all transcription start site RNAs (TSS RNAs) and transcrip-
astrocytes and
the cell types required for the formation of the CNS. tion initiation RNAs (tiRNAs)). These small RNAs range
oligodendrocytes.
Contrary to the earlier dogma that neurogenesis occurs from 18 to 30 nucleotides in length and can shape diverse
only during development, it is now generally accepted cellular pathways (BOX 2). Recent studies have shown that
that neurogenesis occurs throughout life in mammalian several classes of small regulatory RNAs have impor-
brains. Adult neurogenesis has been observed at two tant roles in stem cell biology; indeed, the stem cell and
locations under normal conditions: the subventricular miRNA fields have converged with the identification of
zone (SVZ) of the lateral ventricles and the subgranu- stem cell-specific and lineage-specific miRNAs4,5. It has
lar zone (SGZ) of the dentate gyrus in the hippocampus become clear that miRNAs provide a new dimension to
(FIG. 1). Neurogenesis is regulated at many levels by both the regulation of stem cell functions. For example, owing
extrinsic factors, such as physiological and pathological to their functions in the regulation of translation, miRNAs
conditions (BOX 1; FIG. 1c), and intrinsic factors, such as can regulate stem cell fate and behaviour by fine-tuning
genetic and epigenetic programmes (FIG. 1c). The main- the protein levels of various factors required for stem cell
tenance and differentiation of NSCs are tightly control- proliferation and multipotency. Furthermore, piRNAs
led by intricate molecular networks. Uncovering these are also important for stem cell self-renewal, as their
Department of Human regulatory mechanisms is crucial for understanding the partner Piwi proteins are required for stem cell main-
Genetics, Emory University functions and plasticity of the CNS. tenance. In this Review, we focus on recent progress in
School of Medicine, 615
Genome projects have shown that at least 93% of understanding the roles of small regulatory RNAs
Michael Street, Suite 301,
Atlanta, Georgia 30322, nucleotides in the human genome are transcribed in dif- in neurogenesis.
USA. ferent cells, with similar findings in the mouse and other
Correspondence to P.J. eukaryotes. However, only around 1.2% of transcribed Expression of small RNAs in neurogenesis
e‑mail: peng.jin@emory.edu RNAs encode proteins, indicating that there may be a Detecting nucleic acids that are only 18–30 nucle-
doi:10.1038/nrn2739
Published online
vast reservoir of biologically meaningful non-coding otides in length is inherently difficult, simply because
31 March 2010; corrected RNAs, often referred to as the ‘dark matter’ of the cell3. of their small size. The past several years have seen the
online 30 April 2010 Non-coding RNAs may regulate gene expression at many emergence of new techniques for the identification

NATURE REVIEwS | NeuroscieNce VolUmE 11 | mAy 2010 | 329

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

and profiling of small regulatory RNAs (BOX 3). Earlier progenitor cells, whereas miR-23 is expressed mainly in
expression profiling studies revealed that miRNAs astrocytes. Selective miRNAs are either exclusively or
have spatiotemporal and cell-specific expression preferentially expressed in neurons, including miR-125,
patterns in the brain6–14. The expression of some miRNAs miR-128 and miR-138. Neuronal differentiation of
is brain-specific, such as miR-9, miR-124a, miR-124b embryonic stem (ES) cells induced by retinoic acid sub-
and miR-135. other miRNAs are enriched in the brain, stantially alters the expression profile of miRNAs10,16.
such as let-7, miR-9* (from the opposite arm of the Furthermore, expression of let-7 is highly induced in
miRNA precursor), miR-125a, miR-125b and miR-128 both early neurogenesis and retinoic acid-induced neu-
(ReFs 8–10,13,15) . miRNAs also have cell-specific ronal differentiation in embryocarcinoma, whereas it is
expression patterns. miR-92b is restricted to neuronal decreased in adult brains10,13,16. The specific expression

a Neocortical b
neuroepithelium
LV Blood Type B LV
Neocortex vessel

Type A

Neuronal identity
Neurogenesis
E8 E14 P1 Adult

Neurogenesis-related miRNAs Ependymal


Type C cell

Neuronal specification-related
miRNAs SVZ

Hipp
c CB
OB
Extrinsic factors
• Disease and ageing • Physical activity
• Enriched environment • Hormones
• Growth factors • Neurotransmitters
SGZ

NSC NPC
proliferation differentiation
MCL
Intrinsic factors
Genetics Epigenetics
• Signalling pathways: • DNA methylation
SHH, WNT and Notch • Histone modification GCL
• Transcriptional regulators
• Translational regulators
• Small regulatory RNAs SGZ

Figure 1 | embryonic and adult neurogenesis. a | Mammalian neurogenesis starts around embryonic day 8 (E8), reaches
its peak around E14 and decreases thereafter. The vertebrate CNS is derived from neuroepithelium. For cortical
Nature Reviews | Neuroscience
neurogenesis, neuroepithelial cells begin to acquire features of radial glial cells. These cells serve as the progenitor
population for neuroblasts that migrate to the cortex and differentiate into mature cortical neurons. During neurogenesis,
the expression levels of a subset of microRNAs (miRNAs) increase and are maintained at high levels in neuronal cells,
whereas the expression patterns of other miRNAs mirror the wave of neurogenesis, suggesting that they have different
roles in neurogenesis. b | Adult neurogenesis is observed at two locations under normal conditions: the subventricular
zone (SVZ) of the lateral ventricles (LVs) and the subgranular zone (SGZ) of the dentate gyrus in the hippocampus (Hipp).
Slowly dividing SVZ astrocytes (type B cells) derived from embryonic radial glial cells are neural stem cells (NSCs) that are
relatively quiescent and can give rise to rapidly dividing immature precursors (type C cells). Type C cells actively proliferate
and can generate immature neuroblasts (type A cells), which migrate in chains along the rostral migratory stream to the
olfactory bulb (OB) and differentiate into interneurons. SGZ astrocytes (type B cells) could also function as NSCs. NSCs in
the dentate gyrus undergo proliferation, fate specification, migration, axon and dendrite targeting and synaptic
integration. c | Neurogenesis is regulated at many levels by both extrinsic factors, such as physiological and pathological
conditions, and intrinsic factors, such as genetic and epigenetic programmes. CB, cerebellum; GCL, granular cell layer;
MCL, molecular cell layer; NPC, neuronal progenitor cell; P1, postnatal day 1; SHH, Sonic hedgehog protein.

330 | mAy 2010 | VolUmE 11 www.nature.com/reviews/neuro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Deep sequencing of miRNA in the nervous system strongly suggests that During neurogenesis, neuronal diversity can be
An approach enabled by miRNAs could play key parts in brain development and achieved through the asymmetrical cell division of
next-generation sequencing neuronal fate specification (FIG. 2a). NSCs, which produces two daughter cells with different
technology that is particularly Studies in human and mouse ES cells have shown developmental potentials. whereas one differenti-
useful for identifying low-
abundance RNAs or
that the complete repertoire of miRNAs in stem cells ates into a neuron, the other maintains a stem cell fate.
low-frequency mutations. can only be uncovered through deep sequencing , as Although the mechanism is more complex in verte-
some miRNAs are expressed solely in ES cells. Thus, it brates than invertebrates, the unequal inheritance of cell
Cre–loxP system is important to use this technology to profile the expres- fate determinants (DNA or protein) ensures that each
A site-specific recombination
sion of small regulatory RNAs in both proliferating and daughter cell acquires a different fate23–25. The TRIm-
system derived from
Escherichia coli bacteriophage
differentiated NSCs — an approach that has not been NHl (tripartite motif and NCl1, HT2A and lIN41
P1. Two short DNA sequences taken so far. domain-containing) protein TRIm32, a previously
(loxP sites) are engineered to identified E3 ubiquitin ligase, was recently found to
flank the target DNA. NSC fate determination by miRNAs enhance the activity of selected miRNAs by interacting
Activation of the
Cre-recombinase enzyme
To determine whether miRNAs play a crucial part in with AGo1, a member of the Argonaute protein family
catalyses recombination neurogenesis and the development of the nervous system, involved in miRNA-mediated gene regulation26. During
between the loxP sites, leading early studies focused on key components of the miRNA asymmetrical cell division in NSCs and neural progeni-
to excision of the intervening pathway. loss of Dicer (an RNase III enzyme that tor cells (NPCs), TRIm32 is enriched in one of the two
DNA sequence.
cleaves the miRNA precursor (pre-miRNA) to pro- daughter cells. TRIm32 suppresses cell proliferation and
Environmental enrichment duce mature miRNA) in zebrafish was found to lead to induces neuronal differentiation by degrading the tran-
Providing animals under defective neural tubes, reduced ventricle size and loss scription factor myC and enhancing let-7a activity 26.
managed care with of the midbrain–hindbrain boundary 17. Surprisingly, Because TRIm32 and let-7a are also present in NSCs
environmental stimuli to these defects could be partially rescued by a single and NPCs, it is possible that another pathway might exist
improve the quality of life by
increasing physical activity,
miRNA, miR-430 (ReF. 17). Similarly, the disruption to antagonize the effect of TRIm32–let7a and maintain
stimulating natural behaviours of miRNA-induced silencing complex (miRISC) activity their multipotency 27. These findings suggest that the
and preventing or reducing due to a mutation in Argonaute 2 (AGo2), an essential unequal distribution of a ribonucleoprotein complex-
neural disorders including component of the complex, prevented neural-tube clo- containing TRIm32 and its target miRNAs could regu-
stereotypical behaviours.
sure in mice18. These two studies provide strong in vivo late the balance between differentiation and proliferation
evidence that the miRNA pathway is important for brain in neural stem cell lineages, and thus that miRNAs could
development and morphogenesis. Further advances were participate in cell fate determination and the regulation
provided by studies in which Dicer activity was condi- of asymmetrical cell division.
tionally removed in specific neuronal cell types using Recent studies on individual miRNAs have begun to
the Cre–loxP system. This led to various defects in neural shed light on their specific functions in neurogenesis and
development, such as increased cortical apoptosis, neuronal development (see below)4,28–32. Further research
smaller cortex size, enlarged lateral ventricles, profound in this area could reveal novel mechanisms regulating
astrogliosis, altered dendritic maturation, smaller neural neurogenesis and targets to manipulate this process.
cells and abnormal neurogenesis19–22. These findings
indicate an essential role for the miRNA pathway in Interplay between miRNAs and transcription factors.
neural development. The interplay between miRNAs and transcription
factors was first uncovered in Caenorhabditis elegans.
In C. elegans, two taste neurons, ASE right (ASER) and
Box 1 | Multiple extrinsic factors regulate neurogenesis ASE left (ASEl), display left–right asymmetrical expres-
Diverse intrinsic and extrinsic mechanisms are known to regulate neurogenesis1,2. The sion patterns of putative chemoreceptor genes, which are
following extrinsic factors have been found to be involved: hormones and neural important for functional lateralization of taste neurons.
transmitters, including dopamine, serotonin, acetylcholine and glutamate; growth ASEl and ASER were found to express distinct miRNAs,
factors, such as fibroblast growth factor 2, epidermal growth factor and vascular lsy-6 and miR-273, respectively 33,34. lsy-6 and miR-273
endothelial growth factor; exercise and learning; environmental enrichment and regulate the establishment of neuronal left–right asym-
pathological conditions, such as stress, seizure, ischaemia and ageing. metry by suppressing the translation of their targets,
Mounting evidence indicates the essential role of the stem cell niche in the cog‑1 and die‑1. CoG-1 and DIE-1 can also inhibit
maintenance and regulation of neurogenesis. In the subventricular zone (SVZ) of adult the expression of lsy-6 and miR-273 to form a double-
mice, the cilia of ependymal cells transform the flow of cerebrospinal fluid into a chemical
negative feedback loop33–35. These studies provide an
signal that directs the establishment of the chemorepulsive gradients, which guides the
migration of newly born neurons88. Recent studies revealed that the vascular system is excellent example of how miRNAs can specify distinct
also a key component of the stem cell niche and is involved in regulating neurogenesis in neuronal cell fates from a multipotent precursor state.
the SVZ89,90. Proliferating neural stem cells (NSCs) are tightly apposed to the blood vessels miR-124 is highly conserved in both invertebrates
through the laminin receptor α6β1 integrin, and neuroblast chains run parallel to blood and vertebrates and is the most abundant miRNA in
vessels in the direction of migration towards the rostral migratory stream89,90. both the embryonic and adult CNS8. Its temporal expres-
Vasculature-derived signals can access the SVZ, and most of the proliferating cells that sion pattern suggests it may have a function in neuronal
were recovered from treatment with the anti-mitotic reagent cytosine–arabinofurano- development 7,16. Ectopic expression of miR-124 in cul-
side were directly coupled to the vasculature90, suggesting that the vasculature provides tured non-neuronal cells increased the expression of
signalling cues to NSCs and regulates their function. Given the roles of small regulatory neuronal genes and inhibited non-neuronal genes16,36,37.
RNAs in other types of stem cell niche5, it is likely that they also have important roles in
This is required during early embryonic neurogenesis
the NSC niche, although this remains to be explored.
to ensure that differentiation occurs at the correct

NATURE REVIEwS | NeuroscieNce VolUmE 11 | mAy 2010 | 331

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Transit-amplifying cells time and that the appropriate number of neurons is In the SVZ of the adult mouse, miR-124 shows cell
Cells that arise from adult generated. RE1-silencing transcription factor (REST; type-specific expression. It is expressed in neuroblasts
stem cells and divide a finite also known as NRSF) suppresses neuronal genes in as well as mature granule and periglomerular neu-
number of times until they non-neuronal cells by binding to a conserved repressor rons, but not in dividing doublecortin-negative transit-
become differentiated. They
are committed progenitor cells.
element (RE1) in neuronal gene loci and recruiting the amplifying cells or in oligodendrocytes or astrocytes41.
co-repressor complex containing histone deacetylases During the transition from amplifying type C cells to
and the methyl-CpG-binding protein mECP2 (ReF. 38). neuroblasts, the expression of miR-124 increases and is
Downregulation of REST during the transition from pro- much higher in mature neurons, suggesting miR-124
genitors to post-mitotic neurons allows neuronal gene might be important for cell lineage progression. Further
expression. Small carboxy-terminal domain phosphatase 1 gain- and loss-of-function studies suggest that miR-124
(SCP1; also known as CTDSP1) is another anti-neural regulates the progression of neuronal differentiation by
factor expressed in non-neuronal tissues39. SCP1 can be targeting SoX9, a transcription factor that can maintain
recruited to RE1-containing neuronal genes by REST the pluripotent state of NSCs and inhibit neuronal dif-
in non-neuronal cells. The timely downregulation of ferentiation41 (FIG. 2b). whether SCP1 is regulated by
the anti-neural REST–SCP1 pathway is thought to be miR-124 in adult NSCs remains to be determined. As
important in embryonic neurogenesis. REST binds to well as the interplay between miR-124 and transcrip-
RE1 sites within the promoter regions of three miR-124 tion factors, it has also been shown that miR-124 could
genomic loci and represses their expression through the promote neuronal differentiation and neuronal line-
recruitment of two co-repressor complexes, SIN3 and age commitment by inducing neuron-specific alterna-
CoREST40. miR-124 has been shown to directly target tive splicing of polypyrimidine tract-binding protein 1
the SCP1 3′-untranslated region (UTR), which sup- (PTBP1; also known as PTB and hnRNP I), a global
presses SCP1 expression39. In addition to SCP1, compu- repressor of neuron-specific splicing 37. These findings
tational approaches have also uncovered miR-124 target suggest that miR-124 fine-tunes neurogenesis by target-
sites in the 3′-UTR of mECP2 and CoREST. These ing multiple mRNAs that are involved in different gene
findings suggest the existence of a negative-feedback regulatory mechanisms.
loop between REST–SCP1 and miR-124 that mediates miR-9 is also highly conserved and is expressed in
the rapid transition between neural progenitors and most of the NPCs of zebrafish, chickens and mice6,15,42.
post-mitotic neurons during embryonic neurogenesis39 In Drosophila melanogaster, sensory organs develop
(FIG. 2b). from the divisions of single sensory organ precursor
cells (SoPs). Ectopic miR-9a expression led to a signifi-
cant reduction in the number of SoPs, whereas the loss
of miR-9a resulted in extra production of SoPs. This
Box 2 | Types of small regulatory RNAs and their biogenesis indicates that miR-9a ensures that the precise number of
With the advance of high-throughput sequencing technology, the field of small neuronal precursor cells are generated by controlling cell
regulatory RNAs is rapidly expanding. Currently, the small regulatory RNAs identified fate specification43. Appropriate expression of the tran-
in mammals include microRNAs (miRNAs), endogenous small interfering RNAs (esiRNAs), scription factor Senseless (SENS) is required to control
Piwi-interacting RNAs (piRNAs) and promoter-associated short RNAs (PASRs; also the formation of the precise number of SoPs: high levels
known as transcription start site RNAs (TSS RNAs) and transcription initiation RNAs of SENS in SoPs promote pro-neural gene transcrip-
(tiRNAs))75,83,91,92.
tion, whereas low levels of SENS in non-SoPs repress the
miRNAs are 18–25-nucleotide non-coding RNAs that can regulate the translation of
expression of pro-neural genes. miR-9a inhibits neuronal
target mRNA molecules in a sequence-specific manner. Most endogenous miRNA
genes are transcribed initially as primary transcripts that contain one or more extended fate in non-SoP cells by repressing SENS expression in
hairpin structures. The nuclear RNase III enzyme Drosha, working with DGCR8, cleaves the embryonic and adult stages43.
both strands near the base of the primary stem loop and yields the precursor miRNA During vertebrate brain development, the midbrain–
(pre-miRNA). After being exported to the cytoplasm by exportin 5 (also known as hindbrain boundary (mHB) functions as an organizing
RANGTP), pre-miRNAs are further cleaved by the RNase III enzyme Dicer into mature centre in the neural tube that is both necessary and suf-
miRNAs, which will be incorporated into the RNA-induced silencing complex. It has ficient for the ordered development of the hindbrain.
been shown that a single miRNA can simultaneously regulate the expression of many The mHB contains a pool of NPCs that contributes
mRNA targets and thereby fine-tune protein expression. Many miRNAs are expressed neurons to the entire mHB. Both gain- and loss-of-
in the nervous system, where they affect numerous neuronal genes.
function studies in zebrafish have shown that miR-9 is
High-throughput sequencing has recently identified three new classes of small
required for the normal generation of new neurons in the
regulatory RNAs: esiRNAs, piRNAs and PASRs. The biogenesis of esiRNAs starts with
either bidirectional transcription or the transcription of an inverted repeat. The mHB by repressing the anti-neurogenic genes her5 and
resulting double-stranded RNA or the hairpin RNA precursor is then processed by the her9 (ReF. 44). moreover, ectopic expression of miR-9 in
components of the miRNA pathway. Although miRNA and esiRNA biogenesis is embryonic mouse cortex through in vivo electroporation
generally Dicer-dependent, piRNAs are generated from a long single-stranded led to premature neuronal differentiation in the cor-
precursor independently of Dicer. piRNAs also differ from miRNAs and siRNAs in that tex, produced more neurons in the ventricular zone by
they are usually 26–31 nucleotides long and associated with Piwi subfamily proteins. repressing the expression of forkhead box protein G1
Most piRNAs map to the repetitive regions of the genome and are thought to control (which has been shown to suppress early cortical cell
the activity of transposons. PASRs map to the 5′ end or promoter region of the fate) and disrupted the migration of new neurons in the
protein-coding genes and are proposed to have a role in the transcription of splice
cortex45. In adult neurogenesis, miR-9 inhibits NSC pro-
variants. Whether these small regulatory RNAs are expressed during neurogenesis
liferation and accelerates neural differentiation by tar-
remains to be determined.
geting the nuclear receptor TlX, an essential regulator

332 | mAy 2010 | VolUmE 11 www.nature.com/reviews/neuro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 3 | Expression profiling of small regulatory RNAs


Diverse methodologies have been used to determine the expression of small regulatory RNAs, including northern blot,
in situ hybridization, microarray hybridization-based detection (using locked nucleic acid (LNA) and DNA), reverse
transcription PCR-based detection (TaqMan and SYBR Green), direct sequencing of adaptor-ligated cDNA that is
complementary to the small RNA and solution hybridization to oligonucleotide (LNA and DNA)-conjugated beads.
In recent years, next-generation sequencing technology has been widely used to profile small regulatory RNAs. Currently,
three high-throughput sequencing platforms are commercially available: the Illumina Solexa Genome Analyser, the Roche
454 GS FLX Genome Analyser and Applied Biosystems’s SOLiD system93–95. All three systems require similar preparation of a
small RNA library, including small RNA isolation, reverse transcription and ligation with system-specific adaptors. With the
Roche 454 system, cDNAs are mixed with agarose beads and clonally amplified by emulsion PCR. The products are
randomly deposited to a picotitre plate, and each well holds only one bead, which is pyrosequenced. With the Illumina
Solexa platform, single cDNA strands bind to the surface flow cell channels and are amplified in situ by bridge PCR. Using a
sequencing-by-synthesis approach, each sequencing cycle adds a mixture of four modified deoxynucleotides that are
labelled with four different fluorescence colours. By imaging four channels with laser excitation, the identity of the first base
in each cluster is recorded. After removing the fluorescent label, the next cycle is initiated, and the process is repeated until
the full oligomer is sequenced. With the SOLiD platform, cDNA is clonally amplified by emulsion PCR on the surface of
magnetic beads and subsequently deposited onto the surface of a glass slide. Rather than using polymerase,
ligase-mediated sequencing is performed by introducing a primer to the adaptor sequence of cDNA and then ligating a
fluorescently labelled eight-base oligonucleotide to the sequencing primer. After the fluorescence signal is recorded with
a laser scanner, the octamer is cleaved between the fifth and sixth bases (removing the fluorescent group), and the next
round is repeated until the sequence is completed.

of NSC proliferation and self-renewal, by activating signalling pathway components Fgf8, Fgf receptor 1
wNT–β-catenin signalling and repressing the cyclin- and canopy 1 (ReF. 44). These results indicate that miR-9
dependent kinase inhibitor 1A (CDKN1; also known inhibits the Fgf signalling pathway and promotes neuro-
as wAF1 or CIP1)46. miR-9 suppresses TlX expres- genesis by targeting different mRNAs, which act con-
sion through the 3′-UTR of TlX mRNA47. Introducing vergently to negatively delimit the mHB, and suggest a
a TlX expression vector lacking the endogenous TlX mechanism whereby organizer activity and neurogenesis
3′-UTR rescued the proliferation deficiency induced by are spatially coordinated in the mHB. It also illustrates
miR-9 overexpression and reversed the miR-9-induced a common theme in miRNA-mediated gene regulation;
premature differentiation47. Furthermore, TlX binds to namely, that a given miRNA often regulates several com-
the 3′ genomic sequences of miR-9 and inhibits miR-9 ponents within the same biological pathway to fine-tune
expression. Thus, miR-9 and TlX form a feedback loop the output of this pathway.
to regulate the switch between NSC proliferation and The Sonic hedgehog (SHH) signalling pathway acts
differentiation (FIG. 2b). through the Patched 1–Smoothened (Smo) receptor
In summary, these studies show that transcription complex to activate downstream effectors and has a piv-
factors can directly regulate the expression of specific otal function in cell differentiation, survival and prolif-
miRNAs, and specific miRNAs can then target other eration, and in tissue patterning48. For example, during
transcription factors and regulate their expression Xenopus laevis retinal development, blocking the SHH
post-transcriptionally. The result is a transcription factor pathway delayed cell cycle exit and induced the release of
to miRNA to another transcription factor (sometimes the translational inhibition of two homeobox genes, otx2
even of the miRNA itself ) paradigm for regulating and vsx1, which act as key positive regulators of bipolar
neurogenesis. This represents an interesting, evolution- neuron generation49. It was shown that four miRNAs
arily conserved strategy to keep the balance between (miR-129, miR-155, miR-214 and miR-222) concertedly
miRNAs and their transcriptional regulatory pro- bind to the 3′-UTR of otx2 and vsx1 mRNAs, inhibiting
grammes. However, it has been well documented that their translation and thus the generation of bipolar neu-
many transcription factors can coordinate the regulation rons49. Inhibition of these four miRNAs promotes pro-
of a given gene and that one miRNA can regulate many genitor cell commitment to the bipolar cell fate49. The
mRNA targets. Thus, although the current studies focus SHH pathway also controls the proliferation of cerebellar
on one miRNA and one transcription factor at a time, it granule cell progenitors (GCPs), which respond to SHH
is likely that there is a more complex interplay between produced by Purkinje cells. Excessive SHH signalling in
the miRNA network and the transcriptional regulation GCPs induces neoplastic transformation into medullob-
network in regulating neurogenesis. lastoma. Some miRNAs, including miR-125b, miR-324-5p
and miR-326, are abnormally expressed in medullo-
miRNA-mediated regulation of signalling cascades. blastoma, and these miRNAs inhibit medulloblastoma
miR-9 not only controls neurogenesis at the mHB, but cell proliferation by repressing components of the SHH
Locked nucleic acid also regulates the formation of the mHB in embry- pathway, such as GlI1 and Smo50. Conversely, loss of
A modified RNA nucleotide
with high stability, which can be
onic zebrafish despite not being expressed at this site. these miRNAs has been shown to promote GCP differ-
used as a highly sensitive overexpression of miR-9 in the mHB leads to loss of entiation into granule cells50. A recent study showed that
detection probe. the mHB by inhibiting the fibroblast growth factor (Fgf) the miR-17/92 cluster is upregulated by SHH signalling

NATURE REVIEwS | NeuroscieNce VolUmE 11 | mAy 2010 | 333

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

REST photoreceptor cells. Expression is switched when EGFR


a b
SCP1 signalling transiently triggers yAN degradation (FIG. 2c).
NSC NPC Mature neuron
A major question in signal transduction research is how
miR-124
weak or transient activation of signalling pathways can
SOX9
lead to robust and long-term switches in gene expres-
sion. These studies suggest that, during neurogenesis,
miRNAs may act through feedback loops to reinforce
and stabilize changes in gene expression in response to
let-7 miR-92b miR-124a signalling input.
miR-125 miR-124
miR-9 miR-128
miR-138
NSC NPC Neuron Crosstalk between miRNAs and epigenetic regulation.
Epigenetic regulation, including DNA methylation and
histone modification, is known to be involved in the
GFAP+ Nestin+ NeuN+ TLX modulation of stem cell proliferation and differentia-
Nestin+ DCX+ miR-9 tion, including the proliferation and differentiation of
s100β– PSA–NCAM+
NSCs53,54. Recent genome-wide analyses have shown a
role for DNA methylation and chromatin remodelling,
particularly by the polycomb group proteins, in defining
c the properties and regulating the functions of stem
miR-7 ? Target cells55. Epigenetic regulation and chromatin remodel-
PNT-P1 miR-7 gene
genes
ling are also thought to have important roles in neuro-
Target genesis56–58; however, the identification of downstream
YAN targets has proved elusive59–61.
genes
ORF AAAA Functional interactions between small regulatory
EGFR YAN mRNA RNAs and epigenetic mechanisms in gene silencing
were initially demonstrated in plants and yeast 62, and
Figure 2 | role of mirNAs in fine-tuning neurogenesis. a | Distinct microRNA
(miRNA) expression profiles characterize the transition fromNature
neuralReviews
stem cells (NSCs) to recently in mammals63. on one hand, epigenetic regu-
| Neuroscience
neural progenitor cells (NPCs) and neurons. Marker genes for each of these cell types are lation can modulate small RNA expression and, on
listed below the miRNA profiles. b | Schematic illustration of a transcription factor to the other hand, small regulatory RNAs can induce
miRNA to another transcription factor (sometimes even of the miRNA itself) paradigm for epigenetic changes that lead to mitotically heritable
regulating neurogenesis. This negative-feedback loop keeps the balance between transcriptional silencing 62,64. The crosstalk between
miRNAs and their transcriptional regulatory programmes during neurogenesis (the blue epigenetic regulation and miRNAs in brain develop-
triangles indicate the expression levels of transcription factors and miRNAs). ment and function is now beginning to be investigated.
c | Schematic illustration of the feedback loop between miR-7 and its target, YAN. During A neuronal activity-dependent miRNA, miR-132, was
eye development in Drosophila melanogaster, progenitor cells express YAN, which shown to regulate mECP2 expression in neurons65, and
directly inhibits miR-7 transcription. When progenitor cells differentiate into
the level of mECP2-regulated miR-184 was increased
photoreceptor neurons, epidermal growth factor receptor (EGFR) signalling is activated,
which results in the degradation of YAN. Thus, miR-7 expression is initiated and promotes in neurons following depolarization66. mECP2 belongs
photoreceptor neuron differentiation by directly repressing YAN. Transcription activator to a family of DNA methyl-CpG-binding proteins
pointed P1 (PNT-P1) can also activate miR-7 expression and contribute to YAN that interpret DNA methylation as a signal to sup-
repression. This reciprocal feedback ensures mutually exclusive expression of YAN in press gene expression67. De novo mutations in MECP2
progenitor cells and miR-7 in photoreceptor cells. DCX, doublecortin; GFAP, glial cause neurodevelopmental disorders, including
fibrillary acidic protein; NeuN, neuronal nuclei; ORF, open reading frame; PSA–NCAM, Rett’s syndrome67–69. How the loss of functional mECP2
polysialylated neural cell adhesion molecule; REST, RE1-silencing transcription factor; protein leads to neurodevelopmental deficits is still
SCP1, small carboxy-terminal domain phosphatase 1. under investigation.
Some studies are now revealing a link between miRNAs
and epigenetic regulation in neurogenesis. For example,
in medulloblastoma and promotes GCP proliferation51, gliomas retain many features of NPCs, including the
suggesting that a feedback loop might exist here as well. ability to grow as neurospheres in culture, self-renew
This feedback loop has been shown more clearly in and migrate in the brain. moreover, profiling studies
D. melanogaster. In the eye of the fruitfly, expression of have shown that gliomas express many genes that are
miR-7 is activated in retinal progenitor cells as they begin expressed by NPCs. miRNA expression profiling of
to differentiate into photoreceptors52. This process is human glioblastoma specimens revealed a marked
dependent on epidermal growth factor receptor (EGFR) reduction of miR-128 in tumour samples. miR-128 could
signalling, which triggers extracellular signal-regulated reduce glioma cell proliferation by suppressing BmI1, a
kinase-mediated degradation of the transcription factor member of the polycomb repressive complex 70. It will
yAN. In non-stimulated cells, yAN represses miR-7 tran- be interesting to further examine the role of miR-128 in
scription, whereas miR-7 RNA represses yAN protein neurogenesis.
expression in photoreceptors by binding to sequences NSC differentiation into neurons is accompanied by a
within its mRNA 3′-UTR52. This reciprocal negative feed- switch of neuronal progenitor-specific BRG1-associated
back between yAN and miR-7 ensures mutually exclusive factor (npBAF) to neuron-specific BAF (nBAF) through
expression, with yAN in progenitor cells and miR-7 in ATP-dependent chromatin-remodelling mechanisms.

334 | mAy 2010 | VolUmE 11 www.nature.com/reviews/neuro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

The BAF complex in mammals is functionally related Small regulatory RNAs


to the SwI–SNF complex in Saccharomyces cerevisiae
?
and D. melanogaster, which is thought to facilitate tran- ?
scriptional activation of specific genes by antagonizing ?
chromatin-mediated transcriptional repression. npBAF L1 RNA
has two subunits, BAF53A and BAF45A, which are L1 gene
essential for NSC proliferation; BAF53B and BAF45B, Retrotransposition
by contrast, are subunits of nBAF that are crucial for
dendritic development 71. The repression of BAF53A by
miR-9* and miR-124 was found to mediate the switch
between npBAF and nBAF, suggesting that miRNAs
could target chromatin remodelling factors during L1 gene Neuronal gene
neurogenesis71. However, whether there is an epige-
netic circuitry with a feedback regulatory mechanism Altered neuronal
mediated by miRNAs in the regulation of neurogenesis gene expression
remains to be determined.
Neuronal
miRNAs in the specification of glia diversity
Profiling studies have also identified a range of miRNAs
that help to establish the lineage specificity for both Figure 3 | A model of neuronal diversity generated by
L1 retrotransposition. During Nature Reviews
neuronal | Neuroscience
differentiation,
astrocytes and oligodendrocytes. The phosphorylation
long interspersed nuclear (L1) element transcription is
of signal transducer and activator of transcription 3
activated. This allows subsequent retrotransposition of L1
(STAT3) can inhibit the differentiation of NPCs elements into neuronal genes, which could alter the
into neurons and promote differentiation into glia. expression of adjacent neuronal genes and contribute to
Knockdown of miR-124a and miR-9 increased the level neuronal diversity. L1 RNAs could be processed into small
of phosphorylated STAT3, whereas overexpression of regulatory RNAs, which could potentially regulate L1
miR-124a and miR-9 decreased STAT3 expression, transcription and retrotransposition.
suggesting that these miRNAs could have a role in
modulating glial cell fate determination by regulating
the levels of STAT3 (ReF. 16). In addition, the expression are abundant retrotransposons that make up ~17% of the
of miR-23, a miRNA previously implicated in neural human genome. most l1 elements are retrotransposition
specification, was found to be restricted in astrocytes10; defective; nonetheless, there are approximately 100
however, whether miR-23 is involved in the specification full-length l1 elements that are potentially capable of
of astrocytes requires further study. retrotransposition in the diploid genome. Intriguingly,
To delineate the regulation of oligodendrocyte differ- recent data indicate that l1 element retrotransposition
entiation by miRNAs, oligodendrocyte progenitor cells occurs in rodent and human NPCs, suggesting that
(oPCs) and oligodendrocyte lineage cells (olCs) were l1 elements can have an effect on the expression of
isolated based on their differential expression of cell sur- neuronal genes and therefore influence neuronal cell
A2B5 face markers by fluorescence-activated cell sorting72. There fate and contribute to neuronal diversity 77–80 (FIG. 3). In
A cell surface ganglioside are dynamic changes in the expression of 43 miRNAs addition to the piRNAs that have been mapped to l1
epitope expressed in
developing thymic epithelial
during the transition from A2B5-positive oPCs to pre- elements, the bidirectional transcripts derived from the
cells, oligodendrocyte myelinating olCs. Combined expression profiling of l1 5′-UTR can also be processed to siRNAs that sup-
progenitors and miRNA and mRNA revealed that miR-9 is downregulated press retrotransposition81. It is unclear whether these
neuroendocrine cells. during oligodendrocyte differentiation, which inversely piRNAs or l1-derived siRNAs can modulate l1 element
correlates with the expression of its predicted targets, retrotransposition in adult NSCs and thereby modulate
Long interspersed nuclear
(L1) elements particularly peripheral myelin protein 22 (ReF. 72). Recent neurogenesis.
Full-length active L1 elements studies have identified specific miRNAs involved in the PASRs are thought to be involved in the regulation
are ~6 kb long, consisting of a differentiation of oPCs into oligodendrocytes (see Note of gene expression82,83. Although the exact biological
5′-untranslated region that has added in proof). roles of these small RNAs remain to be determined, the
promoter activity, two open
reading frames (encoding a
involvement of small RNA-mediated transcriptional
nucleic acid-binding protein other small regulatory RNAs in neurogenesis regulation in neurogenesis has been established. In this
and an endonuclease), a Besides miRNAs, numerous piRNAs and esiRNAs have case, a nuclear double-stranded small RNA contain-
reverse transcriptase protein been identified in genomes73–75. piRNAs in particular ing the RE1 sequence that can be recognized by REST
and a poly(A) tail.
have been implicated in controlling the mobilization of was found to be expressed in NSCs. This small RNA
Retrotransposon transposable elements in mice and D. melanogaster 76. induced the neuronal differentiation of NSCs by acti-
Genetic elements that can Although they were initially discovered only in repro- vating the expression of RE1-containing genes84. It can
amplify themselves in a ductive systems, mounting evidence suggests that switch the action of REST from a repressor to an acti-
genome through an RNA piRNAs and esiRNAs are present in both germline and vator, possibly through chromatin modification, and
intermediate. They are
ubiquitous components of the
somatic tissues73–75. In mammals, nearly 15% of piRNAs its absence blocks neuronal differentiation, suggesting
DNA of many eukaryotic have been mapped to long interspersed nuclear (L1) that it is necessary and sufficient to direct NSCs to the
organisms. elements (also known as lINE-1 elements). l1 elements neuronal lineage84.

NATURE REVIEwS | NeuroscieNce VolUmE 11 | mAy 2010 | 335

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Table 1 | MicroRNAs (miRNAs) involved in neurogenesis and their mRNA targets


mirNA Function in neurogenesis cells and tissues Targets refs
let-7 Promotes neuronal lineage commitment NSC LIN28 (mouse) 87
Promotes neural tube closure LIN41 (mouse) 96
miR-7 Promotes neuronal lineage commitment Photoreceptor cell YAN (Drosophila melanogaster) 52
miR-9 Inhibits proliferation NSC and NPC Her5 and Her9 (zebrafish) 44
Promotes neuronal lineage commitment ES cell STAT3 (mouse) 16
Inhibits glial lineage commitment Developing telencephalon FOXG1 (mouse) 45
TLX (mouse) 47
BAF53A (mouse) 71
FGF8 and FGFR1 (mouse) 44
PMP22 (rat) 72
miR-9a Inhibits neuronal fate Epithelial cell Senseless (D. melanogaster) 43
miR-17 Inhibits neural differentiation SH-SY5Y cell Unknown 97
miR-23 Promotes glial lineage commitment ES cell and P19 cell Unknown 10
Promotes neuronal differentiation NT2 cell HES1 (human) 98
miR-124 Inhibits NSC and NPC proliferation Chicken neural tube REST–SCP1 (chicken and mouse) 40
Promotes neuronal lineage commitment P19 cell PTBP1 (mouse) 39
NSC and NPC SOX9 (mouse) 37
BAF53A (mouse) 41
71
miR-125(b) Inhibits NSC and NPC proliferation Medulloblastoma cell and GCP GLI1, SMO and LIN28 (mouse and human) 50
Promotes neuronal lineage commitment SH-SY5Y cell Multiple targets (human) 99
miR-128 Inhibits NSC and NPC proliferation Glioblastoma BMI1 (human) 70
miR-132 Regulates homeostasis Neuron MECP2 (rat) 65
miR-200 Promotes neuronal differentiation Olfactory progenitor cells Notch and TGFβ signalling pathways and FOXG1 100
Maintains olfactory progenitor cells (zebrafish and mouse)
miR-279 Promotes neuronal differentiation Sensory organ precursor Nerfin 1 (D. melanogaster) 101
miR-324-5p Inhibits NSC and NPC proliferation Medulloblastoma cell and GCP GLI1 and SMO (mouse and human) 50
miR-326 Inhibits NSC and NPC proliferation Medulloblastoma cell and GCP GLI1 and SMO (mouse and human) 50
ES cell, embryonic stem cell; FGF8, fibroblast growth factor 8; FGFR1, fibroblast growth factor receptor 1; FOXG1, forkhead box protein G1; GCP, granule cell
progenitor; MECP2, methyl-CpG-binding protein 2; NPC, neural progenitor cell; NSC, neural stem cell; PMP22, peripheral myelin protein 22; PTBP1, polypyrimidine
tract-binding protein 1; REST, RE1-silencing transcription factor; SCP1, small carboxy-terminal domain phosphatase 1; SMO, Smoothened; STAT3, signal
transducer and activator of transcription 3; TGFβ, transforming growth factor-β.

Summary and perspectives with the emergence of powerful deep-sequencing


The discovery of different small regulatory RNAs reveals technologies, there is little doubt that discoveries to
a new layer of complexity in the regulation of gene expres- date represent the ‘tip of the iceberg’, with many more
sion28,85,86, and it is becoming increasingly clear that small small regulatory RNAs involved in regulating neuro-
regulatory RNAs are bona fide regulators of neurogenesis. genesis awaiting identification. Eventually, the interplay
Several principles have emerged from these studies. First, between these small RNAs and different gene regula-
small regulatory RNAs usually function as a fine-tuning tory mechanisms will be systematically unravelled.
mechanism, not as an on–off switch, to ensure the proper These discoveries should paint a more complete pic-
progression through each stage of neurogenesis (from NSC ture of the intricate and complex web of regulatory net-
self-renewal, to lineage specification following differentia- works through which small regulatory RNAs regulate
tion, to the integration of new neurons into existing neural neurogenesis.
circuits) and to coordinate their action with other biologi-
cal regulators such as transcription factors, signalling mol- Note added in proof
ecules and epigenetic effectors. Second, several miRNAs Two recent studies have shown that the miRNA path-
can regulate one mRNA, and a single miRNA is capable of way acts as a ‘micro-brake’ on proliferating oPCs and
regulating many mRNA targets, which could be different promotes the switch of oPCs into post-mitotic oligo-
components within the same pathway (TABLe 1). Finally, sev- dendrocytes102,103. Dicer-knockout mice display severe
eral feedback regulatory loops exist between miRNAs and defects in CNS myelination. miR-219 and miR-338 were
their targets, which could ensure the precision of miRNA- found to promote differentiation into oligodendrocytes
mediated gene regulation. This regulatory circuit involves by repressing several mRNA targets, including those
not only the transcription factors, signalling cascades and encoding platelet-derived growth factor, SoX6 and
epigenetic factors discussed above, but also components HES5. These studies provide compelling evidence that
or regulators of the miRNA pathway itself, such as the miRNAs are essential for regulating oligodendrocyte
feedback loop between let-7 and lIN28 (ReF. 87). differentiation and myelination.

336 | mAy 2010 | VolUmE 11 www.nature.com/reviews/neuro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

1. Ming, G. L. & Song, H. Adult neurogenesis in the 27. Loedige, I. & Filipowicz, W. TRIM-NHL proteins take on 52. Li, X. & Carthew, R. W. A microRNA mediates EGF
mammalian central nervous system. Annu. Rev. miRNA regulation. Cell 136, 818–820 (2009). receptor signaling and promotes photoreceptor
Neurosci. 28, 223–250 (2005). 28. Cao, X., Yeo, G., Muotri, A. R., Kuwabara, T. & Gage, F. H. differentiation in the Drosophila eye. Cell 123,
2. Zhao, C., Deng, W. & Gage, F. H. Mechanisms and Noncoding RNAs in the mammalian central nervous 1267–1277 (2005).
functional implications of adult neurogenesis. system. Annu. Rev. Neurosci. 29, 77–103 (2006). This paper was the first to show that miRNAs
Cell 132, 645–660 (2008). 29. Fineberg, S. K., Kosik, K. S. & Davidson, B. L. regulate neuronal fate through EGF receptor
3. Mattick, J. S. The functional genomics of noncoding MicroRNAs potentiate neural development. Neuron signalling.
RNA. Science 309, 1527–1528 (2005). 64, 303–309 (2009). 53. Li, X. & Zhao, X. Epigenetic regulation of mammalian
4. Gangaraju, V. K. & Lin, H. MicroRNAs: key regulators 30. Kosik, K. S. The neuronal microRNA system. Nature stem cells. Stem Cells Dev. 17, 1043–1052 (2008).
of stem cells. Nature Rev. Mol. Cell Biol. 10, 116–125 Rev. Neurosci. 7, 911–920 (2006). 54. Mehler, M. F. Epigenetics and the nervous system.
(2009). 31. Schratt, G. microRNAs at the synapse. Nature Rev. Ann. Neurol. 64, 602–617 (2008).
5. Stadler, B. M. & Ruohola-Baker, H. Small RNAs: Neurosci. 10, 842–849 (2009). 55. Bernstein, B. E., Meissner, A. & Lander, E. S.
keeping stem cells in line. Cell 132, 563–566 (2008). 32. Coolen, M. & Bally-Cuif, L. MicroRNAs in brain The mammalian epigenome. Cell 128, 669–681
6. Kapsimali, M. et al. MicroRNAs show a wide diversity development and physiology. Curr. Opin. Neurobiol. (2007).
of expression profiles in the developing and mature 19, 461–470 (2009). 56. Lim, D. A. et al. Chromatin remodelling factor Mll1 is
central nervous system. Genome Biol. 8, R173 (2007). 33. Chang, S., Johnston, R. J. Jr, Frokjaer-Jensen, C., essential for neurogenesis from postnatal neural stem
7. Krichevsky, A. M., King, K. S., Donahue, C. P., Lockery, S. & Hobert, O. MicroRNAs act sequentially cells. Nature 458, 529–533 (2009).
Khrapko, K. & Kosik, K. S. A microRNA array reveals and asymmetrically to control chemosensory laterality 57. Ma, D. K. et al. Neuronal activity-induced Gadd45b
extensive regulation of microRNAs during brain in the nematode. Nature 430, 785–789 (2004). promotes epigenetic DNA demethylation and adult
development. RNA 9, 1274–1281 (2003). 34. Johnston, R. J. & Hobert, O. A microRNA controlling neurogenesis. Science 323, 1074–1077 (2009).
8. Lagos-Quintana, M. et al. Identification of tissue- left/right neuronal asymmetry in Caenorhabditis 58. Zhao, X. et al. Mice lacking methyl-CpG binding
specific microRNAs from mouse. Curr. Biol. 12, elegans. Nature 426, 845–849 (2003). protein 1 have deficits in adult neurogenesis and
735–739 (2002). References 33 and 34 provide the first evidence hippocampal function. Proc. Natl Acad. Sci. USA 100,
This is the first paper to report the tissue-specific that miRNAs regulate neuronal fate determination. 6777–6782 (2003).
expression of miRNAs in mammals. 35. Johnston, R. J. Jr, Chang, S., Etchberger, J. F., Ortiz, References 56–58 show the key role of epigenetic
9. Sempere, L. F. et al. Expression profiling of C. O. & Hobert, O. MicroRNAs acting in a double- regulation in neurogenesis.
mammalian microRNAs uncovers a subset of brain- negative feedback loop to control a neuronal cell fate 59. Cheng, L. C., Tavazoie, M. & Doetsch, F. Stem cells:
expressed microRNAs with possible roles in murine decision. Proc. Natl Acad. Sci. USA 102, from epigenetics to microRNAs. Neuron 46, 363–367
and human neuronal differentiation. Genome Biol. 5, 12449–12454 (2005). (2005).
R13 (2004). 36. Lim, L. P. et al. Microarray analysis shows that some 60. Hsieh, J. & Gage, F. H. Epigenetic control of neural
10. Smirnova, L. et al. Regulation of miRNA expression microRNAs downregulate large numbers of target stem cell fate. Curr. Opin. Genet. Dev. 14, 461–469
during neural cell specification. Eur. J. Neurosci. 21, mRNAs. Nature 433, 769–773 (2005). (2004).
1469–1477 (2005). 37. Makeyev, E. V., Zhang, J., Carrasco, M. A. & Maniatis, T. 61. Abel, T. & Zukin, R. S. Epigenetic targets of HDAC
11. Lagos-Quintana, M., Rauhut, R., Lendeckel, W. & The microRNA miR-124 promotes neuronal inhibition in neurodegenerative and psychiatric
Tuschl, T. Identification of novel genes coding for small differentiation by triggering brain-specific alternative disorders. Curr. Opin. Pharmacol. 8, 57–64 (2008).
expressed RNAs. Science 294, 853–858 (2001). pre-mRNA splicing. Mol. Cell 27, 435–448 (2007). 62. Tijsterman, M., Ketting, R. F. & Plasterk, R. H. A.
12. Landgraf, P. et al. A mammalian microRNA expression 38. Ballas, N., Grunseich, C., Lu, D. D., Speh, J. C. & The genetics of RNA silencing. Annu. Rev. Genet. 36,
atlas based on small RNA library sequencing. Mandel, G. REST and its corepressors mediate 489–519 (2002).
Cell 129, 1401–1414 (2007). plasticity of neuronal gene chromatin throughout 63. Morris, K. V., Chan, S. W., Jacobsen, S. E. & Looney,
13. Wulczyn, F. G. et al. Post-transcriptional regulation of neurogenesis. Cell 121, 645–657 (2005). D. J. Small interfering RNA-induced transcriptional
the let-7 microRNA during neural cell specification. 39. Visvanathan, J., Lee, S., Lee, B., Lee, J. W. & Lee, S. K. gene silencing in human cells. Science 305,
FASEB J. 21, 415–426 (2007). The microRNA miR-124 antagonizes the anti-neural 1289–1292 (2004).
14. Wienholds, E. & Plasterk, R. H. MicroRNA function in REST/SCP1 pathway during embryonic CNS 64. Kawasaki, H. & Taira, K. Induction of DNA
animal development. FEBS Lett. 579, 5911–5922 development. Genes Dev. 21, 744–749 (2007). methylation and gene silencing by short interfering
(2005). 40. Conaco, C., Otto, S., Han, J. J. & Mandel, G. RNAs in human cells. Nature 431, 211–217
15. Wienholds, E. et al. MicroRNA expression in zebrafish Reciprocal actions of REST and a microRNA promote (2004).
embryonic development. Science 309, 310–311 (2005). neuronal identity. Proc. Natl Acad. Sci. USA 103, 65. Klein, M. E. et al. Homeostatic regulation of MeCP2
16. Krichevsky, A. M., Sonntag, K. C., Isacson, O. & Kosik, 2422–2427 (2006). expression by a CREB-induced microRNA. Nature
K. S. Specific microRNAs modulate embryonic stem 41. Cheng, L. C., Pastrana, E., Tavazoie, M. & Doetsch, F. Neurosci. 10, 1513–1514 (2007).
cell-derived neurogenesis. Stem Cells 24, 857–864 miR-124 regulates adult neurogenesis in the 66. Nomura, T. et al. MeCP2-dependent repression of
(2006). subventricular zone stem cell niche. Nature Neurosci. an imprinted miR-184 released by depolarization.
17. Giraldez, A. J. et al. MicroRNAs regulate brain 12, 399–408 (2009). Hum. Mol. Genet. 17, 1192–1199 (2008).
morphogenesis in zebrafish. Science 308, 833–838 42. Kloosterman, W. P. et al. Cloning and expression of 67. Bird, A. DNA methylation patterns and epigenetic
(2005). new microRNAs from zebrafish. Nucleic Acids Res. 34, memory. Genes Dev. 16, 6–21 (2002).
This study provides the first in vivo evidence that 2558–2569 (2006). 68. Amir, R. E. et al. Rett syndrome is caused by
miRNAs are crucial for brain development. 43. Li, Y., Wang, F., Lee, J. A. & Gao, F. B. MicroRNA-9a mutations in X-linked MECP2, encoding
18. Liu, J. et al. Argonaute2 is the catalytic engine of ensures the precise specification of sensory organ methyl-CpG-binding protein 2. Nature Genet. 23,
mammalian RNAi. Science 305, 1437–1441 (2004). precursors in Drosophila. Genes Dev. 20, 2793–2805 185–188 (1999).
19. Cuellar, T. L. et al. Dicer loss in striatal neurons (2006). 69. Chahrour, M. & Zoghbi, H. Y. The story of Rett
produces behavioral and neuroanatomical phenotypes 44. Leucht, C. et al. MicroRNA-9 directs late organizer syndrome: from clinic to neurobiology. Neuron 56,
in the absence of neurodegeneration. Proc. Natl Acad. activity of the midbrain–hindbrain boundary. Nature 422–437 (2007).
Sci. USA 105, 5614–5619 (2008). Neurosci. 11, 641–648 (2008). 70. Godlewski, J. et al. Targeting of the Bmi-1 oncogene/
20. Davis, T. H. et al. Conditional loss of Dicer disrupts 45. Shibata, M., Kurokawa, D., Nakao, H., Ohmura, T. & stem cell renewal factor by microRNA-128 inhibits
cellular and tissue morphogenesis in the cortex and Aizawa, S. MicroRNA-9 modulates Cajal-Retzius cell glioma proliferation and self-renewal. Cancer Res. 68,
hippocampus. J. Neurosci. 28, 4322–4330 (2008). differentiation by suppressing Foxg1 expression in 9125–9130 (2008).
21. De Pietri Tonelli, D. et al. miRNAs are essential for mouse medial pallium. J. Neurosci. 28, 71. Yoo, A. S., Staahl, B. T., Chen, L. & Crabtree, G. R.
survival and differentiation of newborn neurons but 10415–10421 (2008). MicroRNA-mediated switching of chromatin-
not for expansion of neural progenitors during early 46. Sun, G., Yu, R. T., Evans, R. M. & Shi, Y. Orphan remodelling complexes in neural development. Nature
neurogenesis in the mouse embryonic neocortex. nuclear receptor TLX recruits histone deacetylases to 460, 642–646 (2009).
Development 135, 3911–3921 (2008). repress transcription and regulate neural stem cell 72. Lau, P. et al. Identification of dynamically regulated
22. Kawase-Koga, Y., Otaegi, G. & Sun, T. Different timings proliferation. Proc. Natl Acad. Sci. USA 104, microRNA and mRNA networks in developing
of Dicer deletion affect neurogenesis and gliogenesis 15282–15287 (2007). oligodendrocytes. J. Neurosci. 28, 11720–11730
in the developing mouse central nervous system. 47. Zhao, C., Sun, G., Li, S. & Shi, Y. A feedback regulatory (2008).
Dev. Dyn. 238, 2800–2812 (2009). loop involving microRNA-9 and nuclear receptor TLX The first paper to report the potential involvement
23. Gotz, M. & Huttner, W. B. The cell biology of in neural stem cell fate determination. Nature Struct. of miRNAs in the development of oligodendrocytes.
neurogenesis. Nature Rev. Mol. Cell Biol. 6, 777–788 Mol. Biol. 16, 365–371 (2009). 73. Kawamura, Y. et al. Drosophila endogenous small
(2005). 48. Ruiz i Altaba, A., Palma, V. & Dahmane, N. RNAs bind to Argonaute 2 in somatic cells. Nature
24. Neumuller, R. A. & Knoblich, J. A. Dividing cellular Hedgehog–Gli signalling and the growth of the brain. 453, 793–797 (2008).
asymmetry: asymmetric cell division and its Nature Rev. Neurosci. 3, 24–33 (2002). 74. Czech, B. et al. An endogenous small interfering RNA
implications for stem cells and cancer. Genes Dev. 23, 49. Decembrini, S. et al. MicroRNAs couple cell fate and pathway in Drosophila. Nature 453, 798–802
2675–2699 (2009). developmental timing in retina. Proc. Natl Acad. Sci. (2008).
25. Zhong, W. & Chia, W. Neurogenesis and asymmetric USA 106, 21179–21184 (2009). 75. Okamura, K. & Lai, E. C. Endogenous small interfering
cell division. Curr. Opin. Neurobiol. 18, 4–11 (2008). 50. Ferretti, E. et al. Concerted microRNA control of RNAs in animals. Nature Rev. Mol. Cell Biol. 9,
26. Schwamborn, J. C., Berezikov, E. & Knoblich, J. A. The Hedgehog signalling in cerebellar neuronal 673–678 (2008).
TRIM-NHL protein TRIM32 activates microRNAs and progenitor and tumour cells. EMBO J. 27, 76. Aravin, A. A., Hannon, G. J. & Brennecke, J. The Piwi-
prevents self-renewal in mouse neural progenitors. 2616–2627 (2008). piRNA pathway provides an adaptive defense in the
Cell 136, 913–925 (2009). 51. Northcott, P. A. et al. The miR-17/92 polycistron is transposon arms race. Science 318, 761–764
This paper was the first to show that a up-regulated in sonic hedgehog-driven (2007).
miRNA-containing ribonucleoprotein complex could medulloblastomas and induced by N-myc in sonic 77. Coufal, N. G. et al. L1 retrotransposition in human
be unequally distributed and contribute to hedgehog-treated cerebellar neural precursors. neural progenitor cells. Nature 460, 1127–1131
asymmetrical cell division. Cancer Res. 69, 3249–3255 (2009). (2009).

NATURE REVIEwS | NeuroscieNce VolUmE 11 | mAy 2010 | 337

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

78. Muotri, A. R. et al. Somatic mosaicism in neuronal 89. Shen, Q. et al. Adult SVZ stem cells lie in a vascular 100. Choi, P. S. et al. Members of the miRNA-200 family
precursor cells mediated by L1 retrotransposition. niche: a quantitative analysis of niche cell-cell regulate olfactory neurogenesis. Neuron 57, 41–55
Nature 435, 903–910 (2005). interactions. Cell Stem Cell 3, 289–300 (2008). (2008).
References 77 and 78 show that 90. Tavazoie, M. et al. A specialized vascular niche for 101. Cayirlioglu, P. et al. Hybrid neurons in a microRNA
retrotransposition of the L1 element occurs adult neural stem cells. Cell Stem Cell 3, 279–288 mutant are putative evolutionary intermediates in
in human and rodent NPCs, which could (2008). insect CO2 sensory systems. Science 319,
contribute to neuronal diversity. References 89 and 90 provide evidence that the 1256–1260 (2008).
79. Kuwabara, T. et al. Wnt-mediated activation of vasculature is an important component of 102. Dugas, J. C. et al. Dicer1 and miR-219 are required for
NeuroD1 and retro-elements during adult neurogenic niches in the adult mammalian brain. normal oligodendrocyte differentiation and
neurogenesis. Nature Neurosci. 12, 1097–1105 91. Ghildiyal, M. & Zamore, P. D. Small silencing RNAs: an myelination. Neuron 65, 597–611 (2010).
(2009). expanding universe. Nature Rev. Genet. 10, 94–108 103. Zhao, X. et al. MicroRNA-mediated control of
80. Muotri, A. R. & Gage, F. H. Generation of neuronal (2009). oligodendrocyte differentiation. Neuron 65, 612–626
variability and complexity. Nature 441, 1087–1093 92. Kim, V. N., Han, J. & Siomi, M. C. Biogenesis of small (2010).
(2006). RNAs in animals. Nature Rev. Mol. Cell Biol. 10,
81. Yang, N. & Kazazian, H. H. Jr. L1 retrotransposition is 126–139 (2009). Acknowledgements
suppressed by endogenously encoded small 93. Mardis, E. R. Next-generation DNA sequencing We thank C. Strauss for critical reading of the manuscript. We
interfering RNAs in human cultured cells. Nature methods. Annu. Rev. Genomics Hum. Genet. 9, apologize to those whose works are not cited here owing
Struct. Mol. Biol. 13, 763–771 (2006). 387–402 (2008). space limitations. The work in our laboratory was supported
82. Kapranov, P. et al. RNA maps reveal new RNA classes 94. Metzker, M. L. Sequencing technologies — the next in part by the grants from the National Institutes of Health
and a possible function for pervasive transcription. generation. Nature Rev. Genet. 11, 31–46 (2010). and International Rett Syndrome Foundation. P.J. is a recipi-
Science 316, 1484–1488 (2007). 95. Rusk, N. & Kiermer, V. Primer: Sequencing — the next ent of the Beckman Young Investigator Award and the Basil
83. Taft, R. J. et al. Tiny RNAs associated with generation. Nature Methods 5, 15 (2008). O’Connor Scholar Research Award, as well as an Alfred P.
transcription start sites in animals. Nature Genet. 41, 96. Maller Schulman, B. R. et al. The let-7 microRNA Sloan Research Fellowship in Neuroscience. X.L. is supported
572–578 (2009). target gene, Mlin41/Trim71 is required for mouse by a FRAXA Fellowship.
84. Kuwabara, T., Hsieh, J., Nakashima, K., Taira, K. & embryonic survival and neural tube closure. Cell Cycle
Gage, F. H. A small modulatory dsRNA specifies the 7, 3935–3942 (2008). Competing interests statement
fate of adult neural stem cells. Cell 116, 779–793 97. Beveridge, N. J., Tooney, P. A., Carroll, A. P., Tran, N. & The authors declare no competing financial interests.
(2004). Cairns, M. J. Down-regulation of miR-17 family
85. Bushati, N. & Cohen, S. M. microRNA functions. expression in response to retinoic acid induced
Annu. Rev. Cell Dev. Biol. 23, 175–205 (2007). neuronal differentiation. Cell Signal. 21, 1837–1845 DATABASES
86. Bartel, D. P. MicroRNAs: target recognition and (2009). OMIM: http://www.ncbi.nlm.nih.gov/omim
regulatory functions. Cell 136, 215–233 (2009). 98. Kawasaki, H. & Taira, K. Hes1 is a target of Rett’s syndrome
87. Rybak, A. et al. A feedback loop comprising lin-28 and microRNA-23 during retinoic-acid-induced neuronal UniProtKB: http://www.uniprot.org
let-7 controls pre-let-7 maturation during neural differentiation of NT2 cells. Nature 423, 838–842 MECP2 | PTBP1 | REST | SCP1 | STAT3 | TLX | TRIM32
stem-cell commitment. Nature Cell Biol. 10, 987–993 (2003).
(2008). 99. Le, M. T. et al. MicroRNA-125b promotes neuronal FuRTHER INFoRMATIoN
88. Sawamoto, K. et al. New neurons follow the flow of differentiation in human cells by repressing Peng Jin’s homepage: http://genetics.emory.edu/labs/jinlab
cerebrospinal fluid in the adult brain. Science 311, multiple targets. Mol. Cell. Biol. 29, 5290–5305 ALL LiNks Are AcTive iN The oNLiNe pdF
629–632 (2006). (2009).

338 | mAy 2010 | VolUmE 11 www.nature.com/reviews/neuro

© 2010 Macmillan Publishers Limited. All rights reserved

You might also like