This article was downloaded by: [University of Phoenix] On: 01 January 2015, At: 15:35 Publisher: Taylor & Francis Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House, 37-41 Mortimer Street, London W1T 3JH, UK

Epigenetics Publication details, including instructions for authors and subscription information: http://www.tandfonline.com/loi/kepi20

Role of miRNAs and epigenetics in neural stem cell fate determination a

Miguel Alejandro Lopez-Ramirez & Stefania Nicoli

a

a

Yale Cardiovascular Research Center; Section of Cardiovascular Medicine; Yale University School of Medicine; New Haven, CT USA Published online: 16 Dec 2013.

Click for updates To cite this article: Miguel Alejandro Lopez-Ramirez & Stefania Nicoli (2014) Role of miRNAs and epigenetics in neural stem cell fate determination, Epigenetics, 9:1, 90-100, DOI: 10.4161/epi.27536 To link to this article: http://dx.doi.org/10.4161/epi.27536

PLEASE SCROLL DOWN FOR ARTICLE Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and should be independently verified with primary sources of information. Taylor and Francis shall not be liable for any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of the Content. This article may be used for research, teaching, and private study purposes. Any substantial or systematic reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http:// www.tandfonline.com/page/terms-and-conditions

Review

Epigenetics 9:1, 90–100; January 2014; © 2014 Landes Bioscience

Role of miRNAs and epigenetics in neural stem cell fate determination Miguel Alejandro Lopez-Ramirez and Stefania Nicoli* Yale Cardiovascular Research Center; Section of Cardiovascular Medicine; Yale University School of Medicine; New Haven, CT USA

Abbreviations: miRNAs, microRNA; CNS, central nervous system; NSC, neural stem cells; 3′UTR, 3′-untranslated region; primiRNA, primary miRNA; DGCR8, DiGeorge syndrome critical region 8, pre-miRNA, precursor miRNA; NE, neuroepithelial; VZ, ventricular zone; SVZ, subventricular zone; SGZ, subgranular zone; RMS, rostral migratory stream; HATs, histone acetyltransferases; HDACs, histone deacetylases; RE1, repressor element-1; REST, RE1 silencing transcription factor; CoREST, C-terminal cofactor for REST; H3K4, histone H3 methylation at lysine 4; Ezh2, enhancer of Zeste homolog 2; LSD1, lysinespecific demethylase A1; DNMT, DNA methyltransferases; MBD, methyl-CpG binding domain; MeCP2, methyl-CpG binding protein 2; MBD1, methyl-CpG binding protein 1; SWI/SNF, switch/sucrose non-fermenting; ISWI, imitation switch; CHD, chromo helicase DNA binding; INO80, inositol auxotroph 80; BAFs, Brg/Brm-associated factors

The regulation of gene expression that determines stem cell fate determination is tightly controlled by both epigenetic and posttranscriptional mechanisms. Indeed, small noncoding RNAs such as microRNAs (miRNAs) are able to regulate neural stem cell fate by targeting chromatin-remodeling pathways. Here, we aim to summarize the latest findings regarding the feedback network of epigenetics and miRNAs during embryonic and adult neurogenesis.

Introduction The cell diversity and high degree of complexity that constitute the developing and adult central nervous system (CNS) are generated by neural stem cells (NSCs). NSCs are subject to neural lineage specification due to changes in gene expression pattern in response to cell-intrinsic mechanisms and cell-extrinsic molecular cues.1,2 Recent studies have shown that epigenetics play an important role in determining the gene expression program of NSCs. Indeed, neural epigenetic control implies heritable changes in gene expression that do not alter the genomic sequence including, histone modifications, DNA methylation, chromatin remodeling and transcriptional feedback loops.2 The epigenetic machinery mediates the switch in the state of chromatin structure and not only provides a platform to recruit binding proteins to allow transcriptional activation or repression, but also confers “bivalent chromatin domains” poised for gene activation.1,3,4 More recently, non-coding RNAs, such as microRNAs (miRNAs), have been included in the epigenetics list, and have been shown to work in concert with the epigenetic machinery in order to regulate NSC fate progression to differentiated progeny.5,6 miRNAs *Correspondence to: Stefania Nicoli; Email: [email protected] Submitted: 10/14/2013; Revised: 12/06/2013; Accepted: 12/16/2013; Published Online: 12/16/2013; http://dx.doi.org/10.4161/epi.27536

are phylogenetically conserved small non-coding RNAs (~22 nt long), and their control of gene expression occurs at the posttranscriptional level by translation inhibition, mRNA decay, or both via specific sequence interactions with the 3′ untranslated region (3′UTR) of the target genes.7,8 miRNA synthesis and maturation follows a stepwise process that is compartmentalized between the nucleus and the cytoplasm. In the nucleus, the primary miRNA transcript (pri-miRNA) is transcribed by RNA polymerase II, resulting in a transcript with a stem-loop structure of approximately 70 nucleotides, a 5′ 7-methylguanylate cap and a 3′-polyadenylated tail.9,10 Drosha, a ribonucleoprotein RNase III enzyme,11 and its cofactor DiGeorge syndrome critical region 8 (DGCR8), a double-stranded RNA binding protein, mediate the release of the stem-loop intermediate known as the precursor miRNA (pre-miRNA).12 The nascent pre-miRNA is then shuttled from the nucleus to the cytoplasm via exportin-5.13 In the cytoplasm, the terminal loop is cleaved from the premiRNA by Dicer, another ribonucleoprotein RNase III enzyme that was first discovered due to its role in processing small interfering RNAs.7 High throughput sequencing experiments have suggested that NSCs possess a specific repertory of miRNAs to modulate their differentiation.14-16 In this review, we will focus on the cellular and molecular mechanisms that miRNAs and the epigenetics machinery coordinate in order to regulate signaling networks during embryonic and adult NSC maintenance and differentiation.

Embryonic and Adult Neurogenesis The mammalian CNS developmental program begins at the neural plate and neural tube, when the neuroepithelial (NE) cells form a continuous layer of cells with apical-basal polarity.17 NE cells are considered the earliest form of NSC, with the potential to give rise to the diversity of neurons and glia cells during development.18,19 At the onset of neurogenesis, NE cells increase the pool of stem cells by undergoing an amplification phase and a fraction of NE cells shift from proliferation to

90 Epigenetics Volume 9 Issue 1

©2014 Landes Bioscience. Do not distribute.

Downloaded by [University of Phoenix] at 15:35 01 January 2015

Keywords: microRNAs, epigenetics machinery, neural stem cells, neurogenesis, cell fate determination

Review

neuron-generating divisions.17,20 In addition, NE cells also give rise to the second type of NSCs, the radial-glia cells, recognized as the major neural progenitor population (Fig. 1).18,21-23 Similar to NE cells, radial-glia cells express the intermediate filament protein nestin, maintain an apical-basal polarity, undergo interkinetic nuclear migration (movement of the nucleus during cell cycle) and their densely packed cell bodies line the apical surface of the pseudostratified ventricular zone (VZ).21-24 However, radial-glia cells show distinctive features that include the expression of several astroglial markers, loss of epithelial tight-junction proteins at the cell-cell junction and show a characteristic cell morphology, in which a long basal process maintains contact with the pial surface while a shorter apical process projects toward the lumen of the VZ (Fig. 1).17,22 Initially, the radialglia basal process serves as a scaffold for the newly born neurons emerging from the VZ in order to migrate toward the cortical plate. Concomitantly radial-glial cells produce intermediate progenitors also known as transit-amplifying cells (Fig. 1).20,24,25 Studies using video time-lapse microscopy in GFP knock-in transgenic mice specific for neurogenic progenitor cells (a type of transit-amplifying cells) revealed that the major sources of neuron-generating cells occurs in the subventricular zone (SVZ) via multiple rounds of cell divisions before the fate switches to terminally differentiated neurons (Fig. 1).20 Moreover, the switch from neurogenesis to gliogenesis occurs in the early neonatal stage.26 Oligodendrocytes are derived from oligodendrocytic progenitor cells produced during early developmental stages, and the radial-glia cells lose their ventricular localization by migration toward the cortical plate where they acquire the multipolar morphology of differentiated astrocyte.21 Adult mammalian neurogenesis retains several of the cellular and molecular processes of neuronal development in embryonic stages (Fig. 1).27,28 However, it is well known that the adult mammalian brain maintains only two neurogenic niches: the SVZ of the lateral ventricles and the subgranular zone (SGZ) in the dentate gyrus of the hippocampus.29,30 The NSCs located in the SVZ (named type B cells) extend basal processes that interact with the brain vasculature to support the adult neurogenic niche.31 Type B cells give origin to the transit-amplifying progenitors (termed type C cells), which proliferate to generate the committed neuroblasts (type A cells) that migrate along the rostral migratory stream (RMS) to reach the olfactory bulb where they differentiate into interneurons (Fig. 1).22,32 In addition, it has also been reported that SVZ type B cells give rise to oligodendrocytes in normal and injured adult brain.33,34 In the neurogenic niche of the SGZ, NSCs (named type 1 cells) present radial glia-like characteristics and give origin to a nonradial precursor known as the type 2 cell, which in turn generates neuroblasts (named type 3 cells) before differentiating into hippocampal granule cells.28,30,35 In contrast to mammals, the adult brains of other animal models, such as zebrafish (Danio rerio), show 16 neurogenic niches distributed all along the rostro-caudal adult brain axis.36 These extensive neurogenic areas confer to the zebrafish brain the ability to fully regenerate after severe traumatic brain lesion.37

Cellular Mechanism of Neural Cell Fate Determination Early studies of neural fate specification in Caenorhabditis elegans and Drosophila melanogaster indicated that cell division patterns play important roles in neural cell fate determination and generation of cell diversity.38,39 Indeed, studies using lineage tracing in mammalian cerebral cortices have shown that both NE cells and radial-glial cells first increase their number by undergoing symmetric proliferative divisions. This mean that one NE cell begets two NE cells or one radial-glia cell gives rise to two radial glia cells.18,40,41 As development proceeds, sequential fate restrictions take place via symmetric42,43 and asymmetric cell divisions in the VZ and SVZ.44 Chenn and McConnell, suggested that the symmetric and asymmetric cell divisions were associated with the cleavage orientation of progenitor cells.45 For instance, cleavage along the vertical plane of the neural progenitors would be more likely to result in two daughter cells that inherit apical cell fate determinants and remain in the VZ to continue cell proliferation (symmetric cell division). Whereas cleavage along the horizontal plane of neural progenitors generate asymmetric cell division that result in apical and basal daughter cells.45 The latter migrates out of the VZ and differentiates into a neuron whereas the former (apical daughter cells) remains attached to the apical VZ.45 Another cellular mechanism that determines the total number of neurons and cell fate determination during neurogenesis is cell cycle regulation.46 Mathematical modeling suggests that a 50% increase in the rate of cell cycle progression in neural progenitors doubles the neuron number during neurogenesis.46,47 In agreement with this observation, neocortical areas show differential regulation of cell cycle kinetics of progenitors that give rise to a different number of neurons that define the anatomical organization and cytoarchitecture of the embryonic cortex.47 Furthermore, cell cycle regulators influence cell fate determination. An increase in the length of the cell cycle leads to a premature switch of NE cells from proliferative to neuron-generating divisions that result in premature neurogenesis in developing mouse embryos.48 It was suggested that lengthening the G1 phase of the NE cell cycle is sufficient to trigger neurogenesis, because even if there is an unequal distribution of determining factors upon mitosis, the cell cycle will be too short, resulting in symmetric daughter cell fates. But, if the cell cycle is long enough, the determining factors are able to induce differentiation resulting in neuron-generating divisions.46,48

Epigenetics Machinery and microRNAs: Molecular Regulators of Neural Cell Fate Program Epigenetics A defining feature of NSCs is their ability to maintain the stem cell population by undergoing self-renewal, and the generation of different neural cells by their multipotent capacity (Fig. 1). Epigenetic mechanisms of gene regulation play a crucial role in these two characteristics. First, the heritable epigenetic code implies establishing a specific chromatin state characterized by specific patterning of histone modifications, which have been

www.landesbioscience.com Epigenetics 91

©2014 Landes Bioscience. Do not distribute.

Downloaded by [University of Phoenix] at 15:35 01 January 2015

Review

©2014 Landes Bioscience. Do not distribute.

Downloaded by [University of Phoenix] at 15:35 01 January 2015

Figure 1. Schematic illustration of miRNAs function in neuronal fate determination during embryonic and adult neurogenesis. Neuroepithelial cells give origin to radial glia, which in turn undergo self-renew via symmetric (not shown) and asymmetric cell divisions (black arrows) in order to amplify the pool of NSCs during embryonic neurogenesis. A variety of neurons are originated from NSCs and intermediate progenitors via sequential fate restrictions regulated by crosstalk between miRNAs, signaling pathways and the epigenetic machinery as indicated by squares. Some radial glia cells are converted into astrocytic stem cells (B cell). B cells give origin to neuronal progenitors (C cell), which are the direct progeny of committed neuroblast (A cell) that differentiate into mature neuron. miRNAs associated with regulation of NSCs maintenance and neuronal commitment are shown in circles. Colored arrows indicate whether the cellular response is increased (↑) or decreased (↓).

92 Epigenetics Volume 9 Issue 1

timing and reduction of neuronal output (Fig. 2).64 Furthermore, mouse embryonic neural progenitors present “bivalent domains” characterized by high levels of both H3K27 trimethylated (H3K27me3) and H3K4me3 at key neurogenic genes. These chromatin domains are silenced but ready to be expressed upon the specific molecular cues.65 Indeed, recruitment of H3K27me3 demethylase activity at the specific loci is essential for bivalent domain resolution during neurogenesis (Fig. 2).65 Another histone demethylase with important roles in regulating NSC proliferations is the lysine-specific demethylase A1 (LSD1),66 which specifically resolves mono- or di-methylation of H3K4 in order to induce gene silencing (Fig. 2).67 DNA methylation DNA methylation occurs at the cytosine residue in CpG dinucleotides and is associated with transcriptional repression. Members of the DNA methyltransferases family (DNMT1, -3a and -3b) establish DNA methylation patterns in the genome during embryogenesis.53,68 Additionally, the DNA methylation pattern can be inherited during symmetric cell divisions in order to maintain the chromatin state that regulates gene expression (Fig. 2). Indeed, methylation of CpG-rich (CpG islands) promoters is recognized as a hallmark of gene repression that facilitates lineage restriction by preventing transcriptional initiation and/ or stabilizing gene silencing.52,68 For example, at the onset of neurogenesis, embryonic NSCs are unable to give rise to macroglia cells, astrocytes and oligodendrocytes, due to DNA hypermethylation at the promoter regions of many astrocyte-specific genes driven by JAK-STAT signaling.69,70 As development proceeds, the promoter regions become demethylated conferring potential to NSCs to differentiate into macroglia cells in response to glia-inducing stimuli.71 In addition, methylated regions are also regulated by the transcriptional repressors methyl-CpG binding domain (MBD) proteins including methyl-CpG binding protein 2 (MeCP2) and methyl-CpG binding protein 1 (MBD1) that recognized methylated CpG sequences (Fig. 2). Mutations of MeCP2 have been associated with the sporadic mental retardation disorder Rett syndrome due to loss of appropriate epigenetic control during neuronal development.72 Moreover, MBD1 is a transcriptional repressor of fibroblast growth factor 2 (Fgf-2),73 and loss of function of MBD1 in mice selectively affects neuronal differentiation and genomic stability, suggesting that DNA methylation might play important roles in genomic stability in the CNS (Fig. 2).73,74 Chromatin remodeling Gene expression is also regulated by the ATP-dependent chromatin remodeling complexes by altering DNA accessibility to transcription factors and to chromatin modifying proteins.75 The ATPase activity of this family of protein complexes is associated with disruption of nucleosome-DNA contacts, movement of nucleosome along DNA and/or exchanges of nucleosomes in order to regulate transcriptional activation and repression (Fig. 2).76-78 The family of ATP-dependent chromatin remodelers is grouped into four types, based on the sequence homology in their ATPase domains: switch/sucrose non-fermenting (SWI/ SNF), imitation switch (ISWI), chromo helicase DNA binding (CHD) and inositol auxotroph 80 (INO80).52 The mammalian

www.landesbioscience.com Epigenetics 93

©2014 Landes Bioscience. Do not distribute.

Downloaded by [University of Phoenix] at 15:35 01 January 2015

shown to specify gene expression patterns, without changes in the DNA sequence, associated with the mechanism of cellular memory in order to maintain the poised nature of NSCs.4,49 Moreover, in early neurogenesis the multipotency of NSC is reduced over time due to changes in the gene expression program associated with specification of neural cell lineages (Fig. 1). Indeed, genes transcribed in earlier progenitors are gradually silenced whereas subsets of cell type-specific genes are turned on, mediated in part by the epigenetic machinery as discussed below (Fig. 2).4 Histone modifications The basic unit of the eukaryotic chromatin is the nucleosome core particle, consisting of superhelical turns of DNA wrapped around an octamer of the core histone proteins (formed by 2 copies of individual histones, H2A, H2B, H3, and H4).50 Histones are subject to posttranslational modification at the N- and C-terminal tails (Fig. 2).51 A series of recent studies have shown that histone acetylation and methylation constitute important molecular mechanisms that regulate gene expression in neural cells.52,53 Histone acetylation is regulated by two groups of enzymes: histone acetyltransferases (HATs) and histone deacetylases (HDACs). HATs promote lysine acetylation at the ε-amino group (NH3 +) that results in removal of the positive charge, thereby relaxing chromatin condensation that is associated with active gene transcription.51,54 Moreover, histone acetylation is a reversible process mediated by HDACs. Histone deacetylation, mediated by HDACs, is linked to transcriptional repression due to an increase in chromatin compaction.50,51 For instance, NSC self-renewal is in part regulated by the transcription factor TLX by directly recruiting HDACs to its downstream target genes, such as the cyclin-dependent kinase inhibitor, p21, and the tumor suppressor gene, PTEN, resulting in the positive regulation of cell cycle progression (Fig. 2).55 Another example of HDAC recruitment in neurogenesis is illustrated by the transcriptional repressor element-1 (RE1) silencing transcription factor (REST), also known as neuron-restrictive silencer factor (NRSF).56,57 In pluripotent stem cells and NSC, REST silences neuronal genes by orchestrating epigenetic mechanism.58,59 Indeed, REST interacts with the RE1 element of target genes and recruits C-terminal cofactor for REST (CoREST) and mSin3A in order to form a co-repressor platforms that recruit HDAC1/2 and histone methyltransferases, which causes chromatin condensation and gene silencing (Fig. 2).60 Histone methylation is associated with transcriptional activation and repression, as well as with bivalent chromatin domains (Fig. 2).51,54,61,62 Indeed, the difference in the functional outcome depends on several factors including the type of histone, and the location and number of methyl groups at the lysine and arginine residue. For instance, histone H3 methylation at lysine 4 (H3K4), H3K36, and H3K79 are linked to gene activation, whereas H3K9 and H3K27, as well as H4K20 and H4K59, have been associated with gene repression.54,63 Recently, it has been reported that the enhancer of Zeste homolog 2 (Ezh2), a H3K27 methyltransferase and component of the polycomb group protein complex, is associated with modulation of NSC cell fate in the cerebral cortex.64 Loss of Ezh2 in progenitor cells results in a marked upregulation of gene expression, changes in the developmental

94 Epigenetics Volume 9 Issue 1

©2014 Landes Bioscience. Do not distribute.

Downloaded by [University of Phoenix] at 15:35 01 January 2015

Figure 2. Molecular mechanisms of the epigenetic-miRNAs regulatory network associated with chromatin remodeling during neurogenesis. (A) Histone modifications are mediated by acetylation, deacetylation and methylation. HATs relax chromatin structure and facilitate transcription, while HDACs increase chromatin condensation and repress transcription. miR-9 and let-7 regulate the expression of the transcription factor TLX that directly recruits HDACs in order to mediate p21 and Pten gene repression in NSCs. miR-137 has emerged as an important regulator of histone methylation regulatory pathway by targeting the demethylase, LSD1, and the H3K27 methyltransferase, Ezh2. miR-137-Ezh2 network might be associated in balancing genespecific bivalent domains in NSCs. 2 (B) DNA methylation is associated with transcriptional repression and negative regulation of several brain-enriched miRNAs. The transcriptional repressor MBD protein, MeCP2, binds to methylated genes in order to silence genes. MeCP2 has been identified as a target of miR-132 in neurons. The transcription factor REST regulates the expression of brain-enriched miR-9/9* and miR-124. miR-9/9* act as a negative feedback regulatory loop on REST silencing complex. (C) ATP-dependent chromatin remodeling complex alter the chromatin structure in order to regulate NSC fate determination. BAF53a expression levels are regulated by miR-124 and miR-9* during the transition between NSCs proliferation to differentiated post-mitotic neuron.122

Crosstalk between Brain-Enriched miRNAs and the Epigenetic Machinery miR-9 miR-9/9* is a highly conserved miRNA abundantly expressed in the CNS of vertebrates.92,93 miR-9 is expressed in neurogenic regions and delimits the midbrain-hindbrain boundary in embryonic vertebrate models, such as Xenopus and zebrafish, by targeting hairy1, her5, her9 and several components of the FGF signaling (fgf8, fgfr1 and canopy1).94,95 Furthermore, miR-9 is expressed in NSCs, neuronal progenitors and some post-mitotic

neurons to dictate pleotropic responses (Fig. 1).95-97 In human neural progenitor cells inhibition of endogenous miR-9 reduces proliferation and promotes cell migration without consequences in neuronal differentiation.96 In contrast, miR-9 decreases TLX, a self-renewal regulator, in adult mouse NSCs that results in negative modulation of proliferation and positive effects on neural cell differentiation.98 Furthermore, gain and loss of function analysis of miR-9 during development of mouse and zebrafish embryos showed that miR-9 suppresses neural progenitor proliferation and promotes neuronal differentiation by modulation of several target genes including Foxg1, Nr2e1, Meis2, and Gsh2.86,94,99 In Drosophila, miR-9a loss and gain of function studies have demonstrated that miR-9a plays important roles in regulating the number of sensory organ precursors (SOPs) by inhibiting the neuronal cell fate in non-SOP cells via silencing of senseless target gene.100 More recently, it has been appreciated that miR-9 is a component of ultradian oscillations that mediate transition from neural proliferative divisions to neural differentiation by dampening Hes1 ultradian oscillations in neural progenitors.101 Furthermore, miR-9 has been suggested to control the ambivalent state of neural progenitors by directly targeting a set of antagonistic genes associated with progenitor proliferation, zic5 and her6, and neuronal differentiation, elavl3.97 All these studies showed that miR-9 acts as a neural regulatory switch at different developmental stages by regulating multiple targets differentially expressed in both spatiotemporal and cellular context-dependent manners.102 Moreover, chromatin immunoprecipitation analysis revealed that REST acts as a regulator of miR-9 genes (Fig. 2).103,104 Indeed, REST inhibits miR-9-2 promoter activity in undifferentiated neuroblastoma cell-line.104 Interestingly, miR-9 and miR-9* (miRNA with similarly sized as miR-9 synthesized from the opposite strand) have a negative feedback loop on REST silencing complex by silencing REST itself and CoREST gene during neural differentiation.104,105 In addition, hypermethylation of CpG islands in the miR-9 gene loci have been associated with various types of cancers indicating the profound control of miRNA expression by epigenetic mechanisms (Fig. 2).6 miR-34a Another miRNA with roles in neuronal fate determination of NSCs is miR-34a. miR-34a is an intergenic miRNA whose promoter harbors CpG islands silenced by hypermethylation in numerous types of cancer (Fig. 2).106 Recently, it was reported that levels of miR-34a expression are under the control of TAp73, a member of the p53 family, which in turn regulates neuronal differentiation via SIRT1 downregulation.107,108 However, miR-34a has also been observed to have negative roles in neuronal differentiation by targeting of Numbl and concomitant increase in Notch signaling (Fig. 1).109 miR-124 miR-124 is temporally upregulated during neural development110,111 and adult neurogenesis.112 Indeed, the expression of miR-124 is very low in neural progenitors but is highly expressed in neuroblast and mature neurons112-114 (Fig. 1). miR-124 has the ability to instruct non-neural cells, such as HeLa cells, to acquire neural gene programs.115 In addition, fibroblast cells transfected with miR-124 alone or in combination with miR-9/9* acquired

www.landesbioscience.com Epigenetics 95

©2014 Landes Bioscience. Do not distribute.

Downloaded by [University of Phoenix] at 15:35 01 January 2015

genome contains 29 members of the SWI2/SNF2-like ATPase family, and two of these, Brg and Brahma (Brm), form part of a chromatin remodeling complex with 10 core subunits BAFs (Brg/ Brm-associated factors).79 Recently, it was shown that the transition from proliferative NSC to differentiated neuron involved switch in subunit composition of npBAF complex. NSC in proliferation requires Brg/Brm-associated factor containing BAF54a and BAF53a subunits, whereas as neural progenitors exit cell cycle the subunits are replaced by the alternative BAF45b, BAF45c and BAF53b subunits in order to establish a post-mitotic neurons (Fig. 2).79 MicroRNAs A number of recent studies have demonstrated the essential role of miRNAs in regulating cellular mechanisms of NSC maintenance and neural commitment (Fig. 1).8,80,81 In addition, miRNAs might also influence the heterogeneity among NSC in the vertebrate nervous system43 by facilitating their transition into new gene expression programs that control their developmental potential and responsiveness to environmental signals. It has been proposed that the cellular miRNA milieu might also be unique in each cell type generated by NSCs and be required to facilitate developmental transitions during neurogenesis and gliogenesis.14-16 Indeed, initial approaches showed that removal of the Dicer gene has serious consequences in the morphogenesis of the CNS in zebrafish.82 However, global Dicer mouse knockouts are lethal at early stages of development.83,84 Our knowledge of key aspects of miRNA involvement in NSC biology was achieved by conditional knockout of the enzyme Dicer and/or individual miRNAs in different neural cells throughout development.85-87 Using these approaches, cell cycle-regulating miRNAs have been identified that regulate self-renewal of neural progenitors (Fig. 1).88,89 Interestingly, experimental evidence suggests that the timing of Dicer deletion during neural development results in different phenotypes and sometimes in contradictory results. For instance, loss of Dicer in the developing mouse cortex using Emx1-Cre did not affect the neural progenitors cell cycle progression, differentiation or viability at E12.5, while defective cortical layering was observed at E13.5 and postnatally.90 In contrast, in the developing retina, loss of Dicer function appears to increase the number of early generated cell types (ganglion cells) and decrease in the production of late-born cells types (rods and Müller cells).91 These differences have been related to the incomplete deletion of Dicer or gradual decay of miRNAs due to differential miRNA stability in these conditional knockouts.

proliferation, whereas endogenous inhibition of miR-137 increases neuronal differentiation in mouse forebrain.1 Additionally, miR137 might have roles in regulating the poised state of adult NSC due to its ability to target Ezh2 (Fig. 2).135 miR-184 miR-184 expression in NSCs is regulated at the epigenetic level by the methyl-CpG binding protein, MBD1.136 Increased levels of miR-184 promote NSC proliferation and inhibit neural lineage progression by targeting Numbl, while the expression of MBD1 represses miR-184 to allow the differentiation of NSC (Fig. 2).136 let-7 let-7 was the first miRNA identified in a wide range of species and tissues including the brain.137 Although the number of let-7 genes varies between organisms138 their mature form is broadly conserved between species.139 Furthermore, members of the let-7 family differ at the 3′ region that might play a role in compensatory target recognition and/or selectivity.140 For instance, let-7i has been shown to inhibit neuronal differentiation by targeting MASH1 and NGN1, two proneural genes,141 while let-7b targets TLX and cyclin D1 that result in reduced progenitor proliferation and increased neuronal differentiation (Fig. 1).142 Recently, let-7c has been reported to be involved in the chronological neurogenesis in Drosophila. Indeed, the cell fate of late-born neurons is regulated by endocrine stimuli that induce the spatial distribution of let-7 expression. The let-7 target gene Abrupt, a transcription factor, modulates the expression levels of the cell adhesion molecule Fasciclin II that plays important roles in neuronal cell identity.143 The diversity in function among let-7 family members has also been observed in different types of cancer cells, where their expression has been found to be under the control of members of DNA methyltransferases by promoting DNA hypermethylation in the promoter region (Fig. 2).6

Conclusions A great effort has been made within the past few years to uncover the gene regulatory network established by chromatin remodeling proteins (epigenetic machinery) and miRNAs during neurogenesis. Indeed, neurogenesis is a process under tight balance and spatio-temporal regulation of gene expression throughout the entire animal’s life. The interplay between epigenetic machinery and miRNAs result in fine-tuning of multiple mRNA transcripts associated with several cellular mechanism of NSCs fate determination, including cell cycle, cell patterning and their role in symmetric and asymmetric cell divisions remains to be fully elucidated. An interesting prospective for the future of these fields will be to uncover whether miRNAs and epigenetics signaling overlap during neuronal cell death such as in aging processes or Alzheimer disease where, both aberrant DNA methylation and miRNAs expression has been observed but not correlated yet. Understanding the epigenetic-miRNA regulatory network might reveal potential therapeutic targets to treat diseases that affect the CNS. Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

96 Epigenetics Volume 9 Issue 1

©2014 Landes Bioscience. Do not distribute.

Downloaded by [University of Phoenix] at 15:35 01 January 2015

a neuron phenotype, suggesting that the combination of these brain-enriched miRNAs have instructive roles in neural fate determination by targeting several pathways.116,117 For example, miR-124 targets the mRNA of PTBP1, a global repressor of alternative splicing,118 laminin γ1 and integrin β1119 in progenitor cells in order to induced neuronal differentiation. Consistent with the role of miR-124 in promoting neuronal differentiation, neural cells missing endogenous miR-124 have increased expression of nonneural genes and loss of neuronal properties.103 Therefore neuronal tissues require high levels of miR-124 in vivo. One of the genes targeted in this process is SCP1, a phosphatase with anti-neural activity.120 In the adult brain, miR-124 induces constitutive and acute neurogenesis by repression of Sox-9112 and by targeting Jag1, one of the major Notch ligands in neurogenic progenitors located in the SVZ.121 Interestingly, similarly to miR-9, miR-124 coordinates and regulates signaling networks and chromatin remodeling to induce neuronal phenotypes. Recently, Yoo and colleagues122 demonstrate that miR-124 in combination with miR-9* regulate the chromatin-remodeling mechanism associated with the switch from neural progenitor proliferation to neuron-generating divisions. Moreover, absence of REST during neuron-generating divisions allows the expression of miR-124 and miR-9*, which in turn represses BAF53a, an ATP-dependent chromatin remodeling protein, in order to promote dendritic outgrowth (Fig. 2).122 miR-132 miR-132 has been shown to play important roles in dendrite maturation and function of newborn neurons in the adult hippocampus.123-125 Mechanistically, miR-132 is rapidly induced by BDNF in order to regulate dendritic morphogenesis by targeting a Rho GTPase protein, p250GAP (Fig. 1).126,127 In addition, MeCP2, an important methyl-CpG binding protein with roles in neuronal maturation, has been identified as a target of miR-132 (Fig. 2).128 Altered expression levels of MeCP2 affect dendritic development and synaptogenesis resulting in neurodevelopmental defects. It has been proposed that MeCP2 expression in neurons is controlled by a feedback loop by which MeCP2 increases BDNF levels that subsequently increases the expression levels of pre-miR-132 (Fig. 2).127 miR-137 miR-137 is expressed in both embryonic and adult vertebrate brains15,129-131 and is found to be enriched in synaptosomes isolated from rat forebrains.132 In vitro, miR-137 levels increase gradually during differentiation of adult NSCs.131,133 It has been suggested that miR-137 induces differentiation of adult mouse NSCs by negative modulation of cell cycle regulation via direct silencing of CDK6 expression (Fig. 1).133 In addition, miR-137 has the capacity to facilitate the transition from embryonic stem cells toward the neuronal fate by targeting transcription factors, KLF4 and TBX3.134 miR-137 forms a regulatory loop with the histone demethylase, LSD1, and the transcription factor, TLX, in order to regulate NSC fate determination.130 Indeed, TLX represses miR-137 expression through the recruitment of LSD1 in NSCs that in turn maintain cell proliferation. Interesting, miR-137 can also inhibit LSD1 expression resulting in decreased NSC proliferation and premature neural differentiation in embryonic mouse brains (Fig. 2).130 Overexpression of miR-137 promotes NSC

Acknowledgments



References 1.

2.

3.

Downloaded by [University of Phoenix] at 15:35 01 January 2015

4.

5.

6.

7.

8.

9.

10.

11.

12.

13.

14.

15.

Hirabayashi Y, Gotoh Y. Epigenetic control of neural precursor cell fate during development. Nat Rev Neurosci 2010; 11:377-88; PMID:20485363; http:// dx.doi.org/10.1038/nrn2810 Ma DK, Marchetto MC, Guo JU, Ming GL, Gage FH, Song H. Epigenetic choreographers of neurogenesis in the adult mammalian brain. Nat Neurosci 2010; 13:1338-44; PMID:20975758; http://dx.doi. org/10.1038/nn.2672 Margueron R, Reinberg D. Chromatin structure and the inheritance of epigenetic information. Nat Rev Genet 2010; 11:285-96; PMID:20300089; http:// dx.doi.org/10.1038/nrg2752 Lunyak VV, Rosenfeld MG. Epigenetic regulation of stem cell fate. Hum Mol Genet 2008; 17(R1):R2836; PMID:18632693; http://dx.doi.org/10.1093/ hmg/ddn149 Jobe EMMA, McQuate AL, Zhao X. Crosstalk among Epigenetic Pathways Regulates Neurogenesis. Front Neurosci 2012; 6:59; PMID:22586361; http:// dx.doi.org/10.3389/fnins.2012.00059 Sato F, Tsuchiya S, Meltzer SJ, Shimizu K. MicroRNAs and epigenetics. FEBS J 2011; 278:1598-609; PMID:21395977; http://dx.doi. org/10.1111/j.1742-4658.2011.08089.x Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004; 116:28197; PMID:14744438; http://dx.doi.org/10.1016/ S0092-8674(04)00045-5 Shi Y, Zhao X, Hsieh J, Wichterle H, Impey S, Banerjee S, Neveu P, Kosik KS. MicroRNA regulation of neural stem cells and neurogenesis. J Neurosci 2010; 30:14931-6; PMID:21068294; http://dx.doi. org/10.1523/JNEUROSCI.4280-10.2010 Lee Y, Kim M, Han J, Yeom KH, Lee S, Baek SH, Kim VN. MicroRNA genes are transcribed by RNA polymerase II. EMBO J 2004; 23:405160; PMID:15372072; http://dx.doi.org/10.1038/ sj.emboj.7600385 Ro S, Park C, Young D, Sanders KM, Yan W. Tissuedependent paired expression of miRNAs. Nucleic Acids Res 2007; 35:5944-53; PMID:17726050; http://dx.doi.org/10.1093/nar/gkm641 Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Rådmark O, Kim S, et al. The nuclear RNase III Drosha initiates microRNA processing. Nature 2003; 425:415-9; PMID:14508493; http:// dx.doi.org/10.1038/nature01957 Han J, Lee Y, Yeom KH, Nam JW, Heo I, Rhee JK, Sohn SY, Cho Y, Zhang BT, Kim VN. Molecular basis for the recognition of primary microRNAs by the Drosha-DGCR8 complex. Cell 2006; 125:887901; PMID:16751099; http://dx.doi.org/10.1016/j. cell.2006.03.043 Yi R, Qin Y, Macara IG, Cullen BR. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev 2003; 17:30116; PMID:14681208; http://dx.doi.org/10.1101/ gad.1158803 Bartel DP, Chen CZ. Micromanagers of gene expression: the potentially widespread influence of metazoan microRNAs. Nat Rev Genet 2004; 5:396-400; PMID:15143321; http://dx.doi.org/10.1038/nrg1328 Jovicic A, Roshan R, Moisoi N, Pradervand S, Moser R, Pillai B, Luthi-Carter R. Comprehensive expression analyses of neural cell-type-specific miRNAs identify new determinants of the specification and maintenance of neuronal phenotypes. J Neurosci 2013; 33:5127-37; PMID:23516279; http://dx.doi. org/10.1523/JNEUROSCI.0600-12.2013

16. He M, Liu Y, Wang X, Zhang MQ, Hannon GJ, Huang ZJ. Cell-type-based analysis of microRNA profiles in the mouse brain. Neuron 2012; 73:3548; PMID:22243745; http://dx.doi.org/10.1016/j. neuron.2011.11.010 17. Götz M, Huttner WB. The cell biology of neurogenesis. Nat Rev Mol Cell Biol 2005; 6:77788; PMID:16314867; http://dx.doi.org/10.1038/ nrm1739 18. Huttner WB, Kosodo Y. Symmetric versus asymmetric cell division during neurogenesis in the developing vertebrate central nervous system. Curr Opin Cell Biol 2005; 17:648-57; PMID:16243506; http:// dx.doi.org/10.1016/j.ceb.2005.10.005 19. Temple S, Qian X. Vertebrate neural progenitor cells: subtypes and regulation. Curr Opin Neurobiol 1996; 6:11-7; PMID:8794050; http://dx.doi.org/10.1016/ S0959-4388(96)80003-1 20. Haubensak W, Attardo A, Denk W, Huttner WB. Neurons arise in the basal neuroepithelium of the early mammalian telencephalon: a major site of neurogenesis. Proc Natl Acad Sci U S A 2004; 101:3196201; PMID:14963232; http://dx.doi.org/10.1073/ pnas.0308600100 21. Kriegstein A, Alvarez-Buylla A. The glial nature of embryonic and adult neural stem cells. Annu Rev Neurosci 2009; 32:149-84; PMID:19555289; http:// dx.doi.org/10.1146/annurev.neuro.051508.135600 22. Malatesta P, Appolloni I, Calzolari F. Radial glia and neural stem cells. Cell Tissue Res 2008; 331:16578; PMID:17846796; http://dx.doi.org/10.1007/ s00441-007-0481-8 23. Götz M, Barde YA. Radial glial cells defined and major intermediates between embryonic stem cells and CNS neurons. Neuron 2005; 46:369-72; PMID:15882633 24. Noctor SC, Flint AC, Weissman TA, Dammerman RS, Kriegstein AR. Neurons derived from radial glial cells establish radial units in neocortex. Nature 2001; 409:714-20; PMID:11217860; http://dx.doi. org/10.1038/35055553 25. Merkle FT, Alvarez-Buylla A. Neural stem cells in mammalian development. Curr Opin Cell Biol 2006; 18:704-9; PMID:17046226; http://dx.doi. org/10.1016/j.ceb.2006.09.008 26. Hirabayashi Y, Gotoh Y. Stage-dependent fate determination of neural precursor cells in mouse forebrain. Neurosci Res 2005; 51:331-6; PMID:15740796; http://dx.doi.org/10.1016/j.neures.2005.01.004 27. Alvarez-Buylla A, Lim DA. For the long run: maintaining germinal niches in the adult brain. Neuron 2004; 41:683-6; PMID:15003168; http://dx.doi. org/10.1016/S0896-6273(04)00111-4 28. Ming GL, Song H. Adult neurogenesis in the mammalian brain: significant answers and significant questions. Neuron 2011; 70:687-702; PMID:21609825; http://dx.doi.org/10.1016/j.neuron.2011.05.001 29. Gage FH. Mammalian neural stem cells. Science 2000; 287:1433-8; PMID:10688783; http://dx.doi. org/10.1126/science.287.5457.1433 30. Doetsch F. A niche for adult neural stem cells. Curr Opin Genet Dev 2003; 13:543-50; PMID:14550422; http://dx.doi.org/10.1016/j.gde.2003.08.012 31. Mirzadeh Z, Merkle FT, Soriano-Navarro M, GarciaVerdugo JM, Alvarez-Buylla A. Neural stem cells confer unique pinwheel architecture to the ventricular surface in neurogenic regions of the adult brain. Cell Stem Cell 2008; 3:265-78; PMID:18786414; http:// dx.doi.org/10.1016/j.stem.2008.07.004

32. Doetsch F, Caillé I, Lim DA, García-Verdugo JM, Alvarez-Buylla A. Subventricular zone astrocytes are neural stem cells in the adult mammalian brain. Cell 1999; 97:703-16; PMID:10380923; http://dx.doi. org/10.1016/S0092-8674(00)80783-7 33. Menn B, Garcia-Verdugo JM, Yaschine C, GonzalezPerez O, Rowitch D, Alvarez-Buylla A. Origin of oligodendrocytes in the subventricular zone of the adult brain. J Neurosci 2006; 26:7907-18; PMID:16870736; http://dx.doi.org/10.1523/ JNEUROSCI.1299-06.2006 34. Nait-Oumesmar B, Decker L, Lachapelle F, AvellanaAdalid V, Bachelin C, Baron-Van Evercooren A. Progenitor cells of the adult mouse subventricular zone proliferate, migrate and differentiate into oligodendrocytes after demyelination. Eur J Neurosci 1999; 11:4357-66; PMID:10594662; http://dx.doi. org/10.1046/j.1460-9568.1999.00873.x 35. Seri B, García-Verdugo JM, Collado-Morente L, McEwen BS, Alvarez-Buylla A. Cell types, lineage, and architecture of the germinal zone in the adult dentate gyrus. J Comp Neurol 2004; 478:35978; PMID:15384070; http://dx.doi.org/10.1002/ cne.20288 36. Grandel H, Kaslin J, Ganz J, Wenzel I, Brand M. Neural stem cells and neurogenesis in the adult zebrafish brain: origin, proliferation dynamics, migration and cell fate. Dev Biol 2006; 295:26377; PMID:16682018; http://dx.doi.org/10.1016/j. ydbio.2006.03.040 37. Kizil C, Kaslin J, Kroehne V, Brand M. Adult neurogenesis and brain regeneration in zebrafish. Dev Neurobiol 2012; 72:429-61; PMID:21595047; http://dx.doi.org/10.1002/dneu.20918 38. Finney M, Ruvkun G. The unc-86 gene product couples cell lineage and cell identity in C. elegans. Cell 1990; 63:895-905; PMID:2257628; http://dx.doi. org/10.1016/0092-8674(90)90493-X 39. Knoblich JA. Mechanisms of asymmetric cell division during animal development. Curr Opin Cell Biol 1997; 9:833-41; PMID:9425348; http://dx.doi. org/10.1016/S0955-0674(97)80085-3 40. Miyata T, Kawaguchi A, Saito K, Kawano M, Muto T, Ogawa M. Asymmetric production of surface-dividing and non-surface-dividing cortical progenitor cells. Development 2004; 131:313345; PMID:15175243; http://dx.doi.org/10.1242/ dev.01173 41. Caviness VS Jr., Takahashi T, Nowakowski RS. Numbers, time and neocortical neuronogenesis: a general developmental and evolutionary model. Trends Neurosci 1995; 18:379-83; PMID:7482802; http://dx.doi.org/10.1016/0166-2236(95)93933-O 42. Delaunay D, Heydon K, Cumano A, Schwab MH, Thomas JL, Suter U, Nave KA, Zalc B, Spassky N. Early neuronal and glial fate restriction of embryonic neural stem cells. J Neurosci 2008; 28:255162; PMID:18322099; http://dx.doi.org/10.1523/ JNEUROSCI.5497-07.2008 43. Franco SJ, Gil-Sanz C, Martinez-Garay I, Espinosa A, Harkins-Perry SR, Ramos C, Müller U. Fate-restricted neural progenitors in the mammalian cerebral cortex. Science 2012; 337:746-9; PMID:22879516; http:// dx.doi.org/10.1126/science.1223616 44. Kowalczyk T, Pontious A, Englund C, Daza RA, Bedogni F, Hodge R, Attardo A, Bell C, Huttner WB, Hevner RF. Intermediate neuronal progenitors (basal progenitors) produce pyramidal-projection neurons for all layers of cerebral cortex. Cereb Cortex 2009; 19:2439-50; PMID:19168665; http://dx.doi. org/10.1093/cercor/bhn260

www.landesbioscience.com Epigenetics 97

©2014 Landes Bioscience. Do not distribute.

The authors are grateful to Anand Narayanan, Catherine Hare, Guillermina Hill-Teran, Emma Ristori and Corey Smith for their critical reading of this manuscript.

61. Burgold T, Spreafico F, De Santa F, Totaro MG, Prosperini E, Natoli G, Testa G. The histone H3 lysine 27-specific demethylase Jmjd3 is required for neural commitment. PLoS One 2008; 3:e3034; PMID:18716661; http://dx.doi.org/10.1371/journal. pone.0003034 62. Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, Fry B, Meissner A, Wernig M, Plath K, et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 2006; 125:315-26; PMID:16630819; http:// dx.doi.org/10.1016/j.cell.2006.02.041 63. Martin C, Zhang Y. The diverse functions of histone lysine methylation. Nat Rev Mol Cell Biol 2005; 6:838-49; PMID:16261189; http://dx.doi. org/10.1038/nrm1761 64. Pereira JD, Sansom SN, Smith J, Dobenecker MW, Tarakhovsky A, Livesey FJ. Ezh2, the histone methyltransferase of PRC2, regulates the balance between self-renewal and differentiation in the cerebral cortex. Proc Natl Acad Sci U S A 2010; 107:1595762; PMID:20798045; http://dx.doi.org/10.1073/ pnas.1002530107 65. Lim DA, Huang YC, Swigut T, Mirick AL, GarciaVerdugo JM, Wysocka J, Ernst P, Alvarez-Buylla A. Chromatin remodelling factor Mll1 is essential for neurogenesis from postnatal neural stem cells. Nature 2009; 458:529-33; PMID:19212323; http://dx.doi. org/10.1038/nature07726 66. Sun G, Alzayady K, Stewart R, Ye P, Yang S, Li W, Shi Y. Histone demethylase LSD1 regulates neural stem cell proliferation. Mol Cell Biol 2010; 30:19972005; PMID:20123967; http://dx.doi.org/10.1128/ MCB.01116-09 67. Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, Casero RA, Shi Y. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 2004; 119:941-53; PMID:15620353; http://dx.doi.org/10.1016/j.cell.2004.12.012 68. Jones PA, Takai D. The role of DNA methylation in mammalian epigenetics. Science 2001; 293:106870; PMID:11498573; http://dx.doi.org/10.1126/ science.1063852 69. Takizawa T, Nakashima K, Namihira M, Ochiai W, Uemura A, Yanagisawa M, Fujita N, Nakao M, Taga T. DNA methylation is a critical cell-intrinsic determinant of astrocyte differentiation in the fetal brain. Dev Cell 2001; 1:749-58; PMID:11740937; http:// dx.doi.org/10.1016/S1534-5807(01)00101-0 70. Fan G, Martinowich K, Chin MH, He F, Fouse SD, Hutnick L, Hattori D, Ge W, Shen Y, Wu H, et al. DNA methylation controls the timing of astrogliogenesis through regulation of JAK-STAT signaling. Development 2005; 132:3345-56; PMID:16014513; http://dx.doi.org/10.1242/dev.01912 71. Kohyama J, Kojima T, Takatsuka E, Yamashita T, Namiki J, Hsieh J, Gage FH, Namihira M, Okano H, Sawamoto K, et al. Epigenetic regulation of neural cell differentiation plasticity in the adult mammalian brain. Proc Natl Acad Sci U S A 2008; 105:180127; PMID:19004774; http://dx.doi.org/10.1073/ pnas.0808417105 72. Robertson KD, Wolffe AP. DNA methylation in health and disease. Nat Rev Genet 2000; 1:11-9; PMID:11262868; http://dx.doi. org/10.1038/35049533 73. Li X, Barkho BZ, Luo Y, Smrt RD, Santistevan NJ, Liu C, Kuwabara T, Gage FH, Zhao X. Epigenetic regulation of the stem cell mitogen Fgf-2 by Mbd1 in adult neural stem/progenitor cells. J Biol Chem 2008; 283:27644-52; PMID:18689796; http://dx.doi. org/10.1074/jbc.M804899200 74. Zhao X, Ueba T, Christie BR, Barkho B, McConnell MJ, Nakashima K, Lein ES, Eadie BD, Willhoite AR, Muotri AR, et al. Mice lacking methyl-CpG binding protein 1 have deficits in adult neurogenesis and hippocampal function. Proc Natl Acad Sci U S A 2003; 100:6777-82; PMID:12748381; http://dx.doi. org/10.1073/pnas.1131928100

75. Hargreaves DC, Crabtree GR. ATP-dependent chromatin remodeling: genetics, genomics and mechanisms. Cell Res 2011; 21:396-420; PMID:21358755; http://dx.doi.org/10.1038/cr.2011.32 76. Varga-Weisz PD, Blank TA, Becker PB. Energydependent chromatin accessibility and nucleosome mobility in a cell-free system. EMBO J 1995; 14:2209-16; PMID:7774579 77. Aoyagi S, Hayes JJ. hSWI/SNF-catalyzed nucleosome sliding does not occur solely via a twistdiffusion mechanism. Mol Cell Biol 2002; 22:7484-90; PMID:12370295; http://dx.doi. org/10.1128/MCB.22.21.7484-7490.2002 78. Strohner R, Wachsmuth M, Dachauer K, Mazurkiewicz J, Hochstatter J, Rippe K, Längst G. A ‘loop recapture’ mechanism for ACF-dependent nucleosome remodeling. Nat Struct Mol Biol 2005; 12:683-90; PMID:16025127; http://dx.doi. org/10.1038/nsmb966 79. Lessard J, Wu JI, Ranish JA, Wan M, Winslow MM, Staahl BT, Wu H, Aebersold R, Graef IA, Crabtree GR. An essential switch in subunit composition of a chromatin remodeling complex during neural development. Neuron 2007; 55:201-15; PMID:17640523; http://dx.doi.org/10.1016/j.neuron.2007.06.019 80. Sun AX, Crabtree GR, Yoo AS. MicroRNAs: regulators of neuronal fate. Curr Opin Cell Biol 2013; 25:215-21; PMID:23374323; http://dx.doi. org/10.1016/j.ceb.2012.12.007 81. Kawahara H, Imai T, Okano H. MicroRNAs in Neural Stem Cells and Neurogenesis. Front Neurosci 2012; 6:30; PMID:22416227; http://dx.doi. org/10.3389/fnins.2012.00030 82. Giraldez AJ, Cinalli RM, Glasner ME, Enright AJ, Thomson JM, Baskerville S, Hammond SM, Bartel DP, Schier AF. MicroRNAs regulate brain morphogenesis in zebrafish. Science 2005; 308:8338; PMID:15774722; http://dx.doi.org/10.1126/ science.1109020 83. Bernstein E, Kim SY, Carmell MA, Murchison EP, Alcorn H, Li MZ, Mills AA, Elledge SJ, Anderson KV, Hannon GJ. Dicer is essential for mouse development. Nat Genet 2003; 35:215-7; PMID:14528307; http://dx.doi.org/10.1038/ng1253 84. Wienholds E, Koudijs MJ, van Eeden FJ, Cuppen E, Plasterk RH. The microRNA-producing enzyme Dicer1 is essential for zebrafish development. Nat Genet 2003; 35:217-8; PMID:14528306; http:// dx.doi.org/10.1038/ng1251 85. Volvert ML, Rogister F, Moonen G, Malgrange B, Nguyen L. MicroRNAs tune cerebral cortical neurogenesis. Cell Death Differ 2012; 19:157381; PMID:22858543; http://dx.doi.org/10.1038/ cdd.2012.96 86. Shibata M, Nakao H, Kiyonari H, Abe T, Aizawa S. MicroRNA-9 regulates neurogenesis in mouse telencephalon by targeting multiple transcription factors. J Neurosci 2011; 31:3407-22; PMID:21368052; http:// dx.doi.org/10.1523/JNEUROSCI.5085-10.2011 87. Sanuki R, Onishi A, Koike C, Muramatsu R, Watanabe S, Muranishi Y, Irie S, Uneo S, Koyasu T, Matsui R, et al. miR-124a is required for hippocampal axogenesis and retinal cone survival through Lhx2 suppression. Nat Neurosci 2011; 14:112534; PMID:21857657; http://dx.doi.org/10.1038/ nn.2897 88. Palm T, Hemmer K, Winter J, Fricke IB, Tarbashevich K, Sadeghi Shakib F, Rudolph IM, Hillje AL, De Luca P, Bahnassawy L, et al. A systemic transcriptome analysis reveals the regulation of neural stem cell maintenance by an E2F1-miRNA feedback loop. Nucleic Acids Res 2013; 41:3699-712; PMID:23396440; http://dx.doi.org/10.1093/nar/ gkt070 89. Wang Y, Blelloch R. Cell cycle regulation by microRNAs in stem cells. Results Probl Cell Differ 2011; 53:459-72; PMID:21630156; http://dx.doi. org/10.1007/978-3-642-19065-0_19

98 Epigenetics Volume 9 Issue 1

©2014 Landes Bioscience. Do not distribute.

Downloaded by [University of Phoenix] at 15:35 01 January 2015

45. Chenn A, McConnell SK. Cleavage orientation and the asymmetric inheritance of Notch1 immunoreactivity in mammalian neurogenesis. Cell 1995; 82:631-41; PMID:7664342; http://dx.doi. org/10.1016/0092-8674(95)90035-7 46. Dehay C, Kennedy H. Cell-cycle control and cortical development. Nat Rev Neurosci 2007; 8:43850; PMID:17514197; http://dx.doi.org/10.1038/ nrn2097 47. Lange C, Huttner WB, Calegari F. Cdk4/cyclinD1 overexpression in neural stem cells shortens G1, delays neurogenesis, and promotes the generation and expansion of basal progenitors. Cell Stem Cell 2009; 5:320-31; PMID:19733543; http://dx.doi. org/10.1016/j.stem.2009.05.026 48. Calegari F, Huttner WB. An inhibition of cyclindependent kinases that lengthens, but does not arrest, neuroepithelial cell cycle induces premature neurogenesis. J Cell Sci 2003; 116:4947-55; PMID:14625388; http://dx.doi.org/10.1242/ jcs.00825 49. Turner BM. Defining an epigenetic code. Nat Cell Biol 2007; 9:2-6; PMID:17199124; http://dx.doi. org/10.1038/ncb0107-2 50. Jenuwein T, Allis CD. Translating the histone code. Science 2001; 293:1074-80; PMID:11498575; http://dx.doi.org/10.1126/science.1063127 51. Kouzarides T. Chromatin modifications and their function. Cell 2007; 128:693-705; PMID:17320507; http://dx.doi.org/10.1016/j.cell.2007.02.005 52. Hashino ASaE. Epigenetic Regulation of Neural Differentiation from Embryonic Stem Cells. Trends in Cell Signaling Pathways in Neuronal Fate Decision 2013. 53. Namihira M, Kohyama J, Abematsu M, Nakashima K. Epigenetic mechanisms regulating fate specification of neural stem cells. Philos Trans R Soc Lond B Biol Sci 2008; 363:2099-109; PMID:18375376; http://dx.doi.org/10.1098/rstb.2008.2262 54. Peterson CL, Laniel MA. Histones and histone modifications. Curr Biol 2004; 14:R546-51; PMID:15268870; http://dx.doi.org/10.1016/j. cub.2004.07.007 55. Sun G, Yu RT, Evans RM, Shi Y. Orphan nuclear receptor TLX recruits histone deacetylases to repress transcription and regulate neural stem cell proliferation. Proc Natl Acad Sci U S A 2007; 104:152827; PMID:17873065; http://dx.doi.org/10.1073/ pnas.0704089104 56. Chong JA, Tapia-Ramírez J, Kim S, Toledo-Aral JJ, Zheng Y, Boutros MC, Altshuller YM, Frohman MA, Kraner SD, Mandel G. REST: a mammalian silencer protein that restricts sodium channel gene expression to neurons. Cell 1995; 80:949-57; PMID:7697725; http://dx.doi.org/10.1016/0092-8674(95)90298-8 57. Schoenherr CJ, Anderson DJ. The neuron-restrictive silencer factor (NRSF): a coordinate repressor of multiple neuron-specific genes. Science 1995; 267:13603; PMID:7871435; http://dx.doi.org/10.1126/ science.7871435 58. Soldati C, Bithell A, Johnston C, Wong KY, Teng SW, Beglopoulos V, Stanton LW, Buckley NJ. Repressor element 1 silencing transcription factor couples loss of pluripotency with neural induction and neural differentiation. Stem Cells 2012; 30:42534; PMID:22162260; http://dx.doi.org/10.1002/ stem.1004 59. Mandel G, Fiondella CG, Covey MV, Lu DD, Loturco JJ, Ballas N. Repressor element 1 silencing transcription factor (REST) controls radial migration and temporal neuronal specification during neocortical development. Proc Natl Acad Sci U S A 2011; 108:16789-94; PMID:21921234; http://dx.doi. org/10.1073/pnas.1113486108 60. Ballas N, Mandel G. The many faces of REST oversee epigenetic programming of neuronal genes. Curr Opin Neurobiol 2005; 15:500-6; PMID:16150588; http://dx.doi.org/10.1016/j.conb.2005.08.015

104. Laneve P, Gioia U, Andriotto A, Moretti F, Bozzoni I, Caffarelli E. A minicircuitry involving REST and CREB controls miR-9-2 expression during human neuronal differentiation. Nucleic Acids Res 2010; 38:6895-905; PMID:20624818; http://dx.doi. org/10.1093/nar/gkq604 105. Packer AN, Xing Y, Harper SQ, Jones L, Davidson BL. The bifunctional microRNA miR-9/miR-9* regulates REST and CoREST and is downregulated in Huntington’s disease. J Neurosci 2008; 28:143416; PMID:19118166; http://dx.doi.org/10.1523/ JNEUROSCI.2390-08.2008 106. Lodygin D, Tarasov V, Epanchintsev A, Berking C, Knyazeva T, Körner H, Knyazev P, Diebold J, Hermeking H. Inactivation of miR-34a by aberrant CpG methylation in multiple types of cancer. Cell Cycle 2008; 7:2591-600; PMID:18719384; http:// dx.doi.org/10.4161/cc.7.16.6533 107. Aranha MM, Santos DM, Solá S, Steer CJ, Rodrigues CM. miR-34a regulates mouse neural stem cell differentiation. PLoS One 2011; 6:e21396; PMID:21857907; http://dx.doi.org/10.1371/journal. pone.0021396 108. Agostini M, Tucci P, Steinert JR, Shalom-Feuerstein R, Rouleau M, Aberdam D, Forsythe ID, Young KW, Ventura A, Concepcion CP, et al. microRNA-34a regulates neurite outgrowth, spinal morphology, and function. Proc Natl Acad Sci U S A 2011; 108:21099104; PMID:22160706; http://dx.doi.org/10.1073/ pnas.1112063108 109. Fineberg SK, Datta P, Stein CS, Davidson BL. MiR34a represses Numbl in murine neural progenitor cells and antagonizes neuronal differentiation. PLoS One 2012; 7:e38562; PMID:22701667; http:// dx.doi.org/10.1371/journal.pone.0038562 110. Maiorano NA, Mallamaci A. Promotion of embryonic cortico-cerebral neuronogenesis by miR-124. Neural Dev 2009; 4:40; PMID:19883498; http:// dx.doi.org/10.1186/1749-8104-4-40 111. Smirnova L, Gräfe A, Seiler A, Schumacher S, Nitsch R, Wulczyn FG. Regulation of miRNA expression during neural cell specification. Eur J Neurosci 2005; 21:1469-77; PMID:15845075; http://dx.doi. org/10.1111/j.1460-9568.2005.03978.x 112. Cheng LC, Pastrana E, Tavazoie M, Doetsch F. miR124 regulates adult neurogenesis in the subventricular zone stem cell niche. Nat Neurosci 2009; 12:399408; PMID:19287386; http://dx.doi.org/10.1038/ nn.2294 113. Deo M, Yu JY, Chung KH, Tippens M, Turner DL. Detection of mammalian microRNA expression by in situ hybridization with RNA oligonucleotides. Dev Dyn 2006; 235:2538-48; PMID:16736490; http:// dx.doi.org/10.1002/dvdy.20847 114. Åkerblom M, Sachdeva R, Barde I, Verp S, Gentner B, Trono D, Jakobsson J. MicroRNA-124 is a subventricular zone neuronal fate determinant. J Neurosci 2012; 32:8879-89; PMID:22745489; http://dx.doi. org/10.1523/JNEUROSCI.0558-12.2012 115. Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM, Castle J, Bartel DP, Linsley PS, Johnson JM. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature 2005; 433:769-73; PMID:15685193; http:// dx.doi.org/10.1038/nature03315 116. Yoo AS, Sun AX, Li L, Shcheglovitov A, Portmann T, Li Y, Lee-Messer C, Dolmetsch RE, Tsien RW, Crabtree GR. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature 2011; 476:22831; PMID:21753754; http://dx.doi.org/10.1038/ nature10323 117. Xue Y, Ouyang K, Huang J, Zhou Y, Ouyang H, Li H, Wang G, Wu Q, Wei C, Bi Y, et al. Direct conversion of fibroblasts to neurons by reprogramming PTBregulated microRNA circuits. Cell 2013; 152:8296; PMID:23313552; http://dx.doi.org/10.1016/j. cell.2012.11.045

118. Makeyev EV, Zhang J, Carrasco MA, Maniatis T. The MicroRNA miR-124 promotes neuronal differentiation by triggering brain-specific alternative pre-mRNA splicing. Mol Cell 2007; 27:435-48; PMID:17679093; http://dx.doi.org/10.1016/j. molcel.2007.07.015 119. Cao X, Pfaff SL, Gage FH. A functional study of miR-124 in the developing neural tube. Genes Dev 2007; 21:531-6; PMID:17344415; http://dx.doi. org/10.1101/gad.1519207 120. Visvanathan J, Lee S, Lee B, Lee JW, Lee SK. The microRNA miR-124 antagonizes the antineural REST/SCP1 pathway during embryonic CNS development. Genes Dev 2007; 21:744-9; PMID:17403776; http://dx.doi.org/10.1101/ gad.1519107 121. Liu XS, Chopp M, Zhang RL, Tao T, Wang XL, Kassis H, Hozeska-Solgot A, Zhang L, Chen C, Zhang ZG. MicroRNA profiling in subventricular zone after stroke: MiR-124a regulates proliferation of neural progenitor cells through Notch signaling pathway. PLoS One 2011; 6:e23461; PMID:21887253; http://dx.doi.org/10.1371/journal.pone.0023461 122. Yoo AS, Staahl BT, Chen L, Crabtree GR. MicroRNA-mediated switching of chromatin-remodelling complexes in neural development. Nature 2009; 460:642-6; PMID:19561591 123. Magill ST, Cambronne XA, Luikart BW, Lioy DT, Leighton BH, Westbrook GL, Mandel G, Goodman RH. microRNA-132 regulates dendritic growth and arborization of newborn neurons in the adult hippocampus. Proc Natl Acad Sci U S A 2010; 107:203827; PMID:21059906; http://dx.doi.org/10.1073/ pnas.1015691107 124. Hansen KF, Sakamoto K, Wayman GA, Impey S, Obrietan K. Transgenic miR132 alters neuronal spine density and impairs novel object recognition memory. PLoS One 2010; 5:e15497; PMID:21124738; http:// dx.doi.org/10.1371/journal.pone.0015497 125. Luikart BW, Bensen AL, Washburn EK, Perederiy JV, Su KG, Li Y, Kernie SG, Parada LF, Westbrook GL. miR-132 mediates the integration of newborn neurons into the adult dentate gyrus. PLoS One 2011; 6:e19077; PMID:21611182; http://dx.doi. org/10.1371/journal.pone.0019077 126. Wayman GA, Davare M, Ando H, Fortin D, Varlamova O, Cheng HY, Marks D, Obrietan K, Soderling TR, Goodman RH, et al. An activityregulated microRNA controls dendritic plasticity by down-regulating p250GAP. Proc Natl Acad Sci U S A 2008; 105:9093-8; PMID:18577589; http://dx.doi. org/10.1073/pnas.0803072105 127. Vo N, Klein ME, Varlamova O, Keller DM, Yamamoto T, Goodman RH, Impey S. A cAMPresponse element binding protein-induced microRNA regulates neuronal morphogenesis. Proc Natl Acad Sci U S A 2005; 102:16426-31; PMID:16260724; http://dx.doi.org/10.1073/pnas.0508448102 128. Klein ME, Lioy DT, Ma L, Impey S, Mandel G, Goodman RH. Homeostatic regulation of MeCP2 expression by a CREB-induced microRNA. Nat Neurosci 2007; 10:1513-4; PMID:17994015; http:// dx.doi.org/10.1038/nn2010 129. Kapsimali M, Kloosterman WP, de Bruijn E, Rosa F, Plasterk RH, Wilson SW. MicroRNAs show a wide diversity of expression profiles in the developing and mature central nervous system. Genome Biol 2007; 8:R173; PMID:17711588; http://dx.doi. org/10.1186/gb-2007-8-8-r173 130. Sun G, Ye P, Murai K, Lang MF, Li S, Zhang H, Li W, Fu C, Yin J, Wang A, et al. miR-137 forms a regulatory loop with nuclear receptor TLX and LSD1 in neural stem cells. Nat Commun 2011; 2:529; PMID:22068596; http://dx.doi.org/10.1038/ ncomms1532

www.landesbioscience.com Epigenetics 99

©2014 Landes Bioscience. Do not distribute.

Downloaded by [University of Phoenix] at 15:35 01 January 2015

90. De Pietri Tonelli D, Pulvers JN, Haffner C, Murchison EP, Hannon GJ, Huttner WB. miRNAs are essential for survival and differentiation of newborn neurons but not for expansion of neural progenitors during early neurogenesis in the mouse embryonic neocortex. Development 2008; 135:391121; PMID:18997113; http://dx.doi.org/10.1242/ dev.025080 91. Georgi SART, Reh TA. Dicer is required for the transition from early to late progenitor state in the developing mouse retina. J Neurosci 2010; 30:404861; PMID:20237275; http://dx.doi.org/10.1523/ JNEUROSCI.4982-09.2010 92. Lagos-Quintana M, Rauhut R, Yalcin A, Meyer J, Lendeckel W, Tuschl T. Identification of tissue-specific microRNAs from mouse. Curr Biol 2002; 12:735-9; PMID:12007417; http://dx.doi. org/10.1016/S0960-9822(02)00809-6 93. Wienholds E, Kloosterman WP, Miska E, AlvarezSaavedra E, Berezikov E, de Bruijn E, Horvitz HR, Kauppinen S, Plasterk RH. MicroRNA expression in zebrafish embryonic development. Science 2005; 309:310-1; PMID:15919954; http://dx.doi. org/10.1126/science.1114519 94. Leucht C, Stigloher C, Wizenmann A, Klafke R, Folchert A, Bally-Cuif L. MicroRNA-9 directs late organizer activity of the midbrain-hindbrain boundary. Nat Neurosci 2008; 11:641-8; PMID:18454145; http://dx.doi.org/10.1038/nn.2115 95. Bonev B, Pisco A, Papalopulu N. MicroRNA-9 reveals regional diversity of neural progenitors along the anterior-posterior axis. Dev Cell 2011; 20:1932; PMID:21238922; http://dx.doi.org/10.1016/j. devcel.2010.11.018 96. Delaloy C, Liu L, Lee JA, Su H, Shen F, Yang GY, Young WL, Ivey KN, Gao FB. MicroRNA-9 coordinates proliferation and migration of human embryonic stem cell-derived neural progenitors. Cell Stem Cell 2010; 6:323-35; PMID:20362537; http:// dx.doi.org/10.1016/j.stem.2010.02.015 97. Coolen M, Thieffry D, Drivenes Ø, Becker TS, Bally-Cuif L. miR-9 controls the timing of neurogenesis through the direct inhibition of antagonistic factors. Dev Cell 2012; 22:1052-64; PMID:22595676; http://dx.doi.org/10.1016/j.devcel.2012.03.003 98. Zhao C, Sun G, Li S, Shi Y. A feedback regulatory loop involving microRNA-9 and nuclear receptor TLX in neural stem cell fate determination. Nat Struct Mol Biol 2009; 16:365-71; PMID:19330006; http://dx.doi.org/10.1038/nsmb.1576 99. Shibata M, Kurokawa D, Nakao H, Ohmura T, Aizawa S. MicroRNA-9 modulates Cajal-Retzius cell differentiation by suppressing Foxg1 expression in mouse medial pallium. J Neurosci 2008; 28:1041521; PMID:18842901; http://dx.doi.org/10.1523/ JNEUROSCI.3219-08.2008 100. Li Y, Wang F, Lee JA, Gao FB. MicroRNA-9a ensures the precise specification of sensory organ precursors in Drosophila. Genes Dev 2006; 20:2793805; PMID:17015424; http://dx.doi.org/10.1101/ gad.1466306 101. Bonev B, Stanley P, Papalopulu N. MicroRNA-9 Modulates Hes1 ultradian oscillations by forming a double-negative feedback loop. Cell reports 2012; 2:10-8. 102. Uchida N. MicroRNA-9 controls a migratory mechanism in human neural progenitor cells. Cell Stem Cell 2010; 6:294-6; PMID:20362531; http://dx.doi. org/10.1016/j.stem.2010.03.010 103. Conaco C, Otto S, Han JJ, Mandel G. Reciprocal actions of REST and a microRNA promote neuronal identity. Proc Natl Acad Sci U S A 2006; 103:24227; PMID:16461918; http://dx.doi.org/10.1073/ pnas.0511041103

135. Szulwach KELX, Li X, Smrt RD, Li Y, Luo Y, Lin L, Santistevan NJ, Li W, Zhao X, Jin P. Cross talk between microRNA and epigenetic regulation in adult neurogenesis. J Cell Biol 2010; 189:12741; PMID:20368621; http://dx.doi.org/10.1083/ jcb.200908151 136. Liu C, Teng ZQ, Santistevan NJ, Szulwach KE, Guo W, Jin P, Zhao X. Epigenetic regulation of miR-184 by MBD1 governs neural stem cell proliferation and differentiation. Cell Stem Cell 2010; 6:43344; PMID:20452318; http://dx.doi.org/10.1016/j. stem.2010.02.017 137. Pasquinelli AE, Reinhart BJ, Slack F, Martindale MQ, Kuroda MI, Maller B, Hayward DC, Ball EE, Degnan B, Müller P, et al. Conservation of the sequence and temporal expression of let-7 heterochronic regulatory RNA. Nature 2000; 408:86-9; PMID:11081512; http://dx.doi.org/10.1038/35040556 138. Roush S, Slack FJ. The let-7 family of microRNAs. Trends Cell Biol 2008; 18:505-16; PMID:18774294; http://dx.doi.org/10.1016/j.tcb.2008.07.007 139. Friedman RCFK, Farh KK, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 2009; 19:92-105; PMID:18955434; http://dx.doi.org/10.1101/ gr.082701.108

140. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 2009; 136:215-33; PMID:19167326; http://dx.doi.org/10.1016/j. cell.2009.01.002 141. Cimadamore F, Amador-Arjona A, Chen C, Huang CT, Terskikh AV. SOX2-LIN28/let-7 pathway regulates proliferation and neurogenesis in neural precursors. Proc Natl Acad Sci U S A 2013; 110:E3017-26; PMID:23884650; http://dx.doi.org/10.1073/ pnas.1220176110 142. Zhao C, Sun G, Li S, Lang MF, Yang S, Li W, Shi Y. MicroRNA let-7b regulates neural stem cell proliferation and differentiation by targeting nuclear receptor TLX signaling. Proc Natl Acad Sci U S A 2010; 107:1876-81; PMID:20133835; http://dx.doi. org/10.1073/pnas.0908750107 143. Kucherenko MM, Barth J, Fiala A, Shcherbata HR. Steroid-induced microRNA let-7 acts as a spatiotemporal code for neuronal cell fate in the developing Drosophila brain. EMBO J 2012; 31:4511-23; PMID:23160410; http://dx.doi.org/10.1038/ emboj.2012.298

100 Epigenetics Volume 9 Issue 1

©2014 Landes Bioscience. Do not distribute.

Downloaded by [University of Phoenix] at 15:35 01 January 2015

131. Smrt RD, Szulwach KE, Pfeiffer RL, Li X, Guo W, Pathania M, Teng ZQ, Luo Y, Peng J, Bordey A, et al. MicroRNA miR-137 regulates neuronal maturation by targeting ubiquitin ligase mind bomb-1. Stem Cells 2010; 28:1060-70; PMID:20506192; http:// dx.doi.org/10.1002/stem.431 132. Siegel G, Obernosterer G, Fiore R, Oehmen M, Bicker S, Christensen M, Khudayberdiev S, Leuschner PF, Busch CJ, Kane C, et al. A functional screen implicates microRNA-138-dependent regulation of the depalmitoylation enzyme APT1 in dendritic spine morphogenesis. Nat Cell Biol 2009; 11:70516; PMID:19465924; http://dx.doi.org/10.1038/ ncb1876 133. Silber J, Lim DA, Petritsch C, Persson AI, Maunakea AK, Yu M, Vandenberg SR, Ginzinger DG, James CD, Costello JF, et al. miR-124 and miR-137 inhibit proliferation of glioblastoma multiforme cells and induce differentiation of brain tumor stem cells. BMC Med 2008; 6:14; PMID:18577219; http:// dx.doi.org/10.1186/1741-7015-6-14 134. Ke Jiang CR, Venugopalan D. Nair. microrna-137 represses Klf4 and Tbx3 during differentiation of mouse embryonic stem cells. Stem Cell Res (Amst) 2013; In press.

Role of miRNAs and epigenetics in neural stem cell fate determination.

The regulation of gene expression that determines stem cell fate determination is tightly controlled by both epigenetic and posttranscriptional mechan...
2MB Sizes 0 Downloads 0 Views