Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Dicer Is Required for Normal Cerebellar Development and to Restrain Medulloblastoma Formation

  • Frederique Zindy,

    Affiliation Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States of America

  • Youngsoo Lee,

    Current address: Genomic Instability Research Center, Ajou University, School of Medicine, Suwon, South Korea

    Affiliation Department of Genetics, St. Jude Children’s Research Hospital, Memphis, TN, United States of America

  • Daisuke Kawauchi,

    Current address: Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany

    Affiliation Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States of America

  • Olivier Ayrault,

    Current address: Institut Curie/CNRS, UMR 3306/INSERM U1005, Centre Universitaire, Orsay, France

    Affiliation Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States of America

  • Leila Ben Merzoug,

    Affiliation Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States of America

  • Yang Li,

    Affiliation Department of Genetics, St. Jude Children’s Research Hospital, Memphis, TN, United States of America

  • Peter J. McKinnon,

    Affiliation Department of Genetics, St. Jude Children’s Research Hospital, Memphis, TN, United States of America

  • Martine F. Roussel

    martine.roussel@stjude.org

    Affiliation Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States of America

Abstract

Dicer, a ribonuclease III enzyme, is required for the maturation of microRNAs. To assess its role in cerebellar and medulloblastoma development, we genetically deleted Dicer in Nestin-positive neural progenitors and in mice lacking one copy for the Sonic Hedgehog receptor, Patched 1. We found that conditional loss of Dicer in mouse neural progenitors induced massive Trp53-independent apoptosis in all proliferative zones of the brain and decreased proliferation of cerebellar granule progenitors at embryonic day 15.5 leading to abnormal cerebellar development and perinatal lethality. Loss of one copy of Dicer significantly accelerated the formation of mouse medulloblastoma of the Sonic Hedgehog subgroup in Patched1-heterozygous mice. We conclude that Dicer is required for proper cerebellar development, and to restrain medulloblastoma formation.

Introduction

MicroRNAs (miRNAs) are short non-coding RNAs derived from pri-miRNAs that are transcribed and processed by Drosha and the microprocessor complex subunit Di George syndrome critical region gene 8 (DGCR8) into pre-miRNAs in the nucleus [1,2]. Pre-miRNAs are then translocated into the cytoplasm to be further processed by Dicer into mature miRNAs. Mature single-stranded miRNAs are loaded into the RNA-induced silencing complex (RISC) that binds the 3’-UTR of mRNAs to inhibit their translation or induce their degradation [1,2]. Dicer is essential for early mouse development since its loss in the germline induces embryonic lethality at E7.5 [3]. However, conditional deletion of Dicer with Cre transgenic lines in different regions of the nervous system, including inner ear [4,5], retina [6,7], cortex and hippocampus [812], Purkinje cells [13], dopaminoceptive neurons [14,15], Schwann cells [16], astrocytes [17] and glia cells [18], reveals a role of Dicer and miRNAs in different developmental processes including cell survival, differentiation and proliferation. In mice, the full loss of Dicer expression is selected against in tumorigenesis [1921], and loss of two copies of Dicer reduces cell proliferation and increases apoptosis [22]. Dicer is haploinsufficient for tumor suppression in some [19,20], but not all cancers [21]. In humans, germline (including DICER1 syndrome) and somatic mutations of one copy of DICER1 has been described in different tumor types, including one human medulloblastoma [19,23,24]. However, loss of heterozygosity is rare, but has been observed in pineoblastoma and pituitary blastoma [24].

Cerebellar granule neuron progenitors (GNPs), born in the rostral rhombic lip (rRL) at embryonic days (E) 11 to E16, migrate on the surface of the developing cerebellum to form the external granule layer (EGL) [25]. After birth, upon Sonic Hedgehog (SHH) stimulation, GNPs rapidly proliferate with maximum proliferation at postnatal days (P) 5 –P7, after which they exit the cell cycle and migrate inward to form the internal granule layer (IGL). In the mouse, the cerebellum is fully formed by 3 weeks after birth [25]. Constitutive activation of the SHH signaling pathway leads to defects in cell cycle exit, migration and differentiation which in turn can induce medulloblastoma.

The miR-17~92 cluster family is expressed in proliferating GNPs [26] and up-regulated in SHH medulloblastoma [2628]. Co-deletion of miR-17~92 with its paralog miR-106b~25 in mice reduces proliferation of GNPs at postnatal age leading to a small cerebellum [29]. While loss of miR-17~92 leads to a relatively mild phenotype, it is absolutely required for SHH medulloblastoma formation [29]. We here show that Dicer that processes pre-miRNAs into mature miRNAs, including those encoded by the miR-17~92 cluster, is required for both normal cerebellar development and medulloblastoma suppression.

Material and Methods

Mouse husbandry

Mouse lines carrying conditional alleles of Dicer (Dicerfloxed/floxed) [30] were generously provided by Dr. Gregory Hannon (CSHL, NY, USA). Trp53-null mice (stock number 002101; [31]) and the transgenic line in which the Cre recombinase is expressed under the promoter of the rat Nestin gene (Nestin-Cre) (stock number 003771; [32]) were obtained from the Jackson Laboratory (Bar Harbor, ME, USA). Patched1-heterozygous (Ptch1+/-) mice in a Cdkn2c-null background (Ptch1+/-; Cdkn2c-/-) were previously described [33]. All mice were maintained on a mixed 129 x C57BL/6 background.

For animals that will develop medulloblastoma, we made every effort to minimize suffering by daily observation for clinical signs of sickness. Humane endpoints included dome head, titled head, loss of greater than 20% body weight, slow movements, difficulties with gait and dehydration. In the Ptch1+/-; Cdkn2c-/- genetic background, ~ 60% of the mice will develop medulloblastoma while the other 40% of animals do not develop tumors. Mice with any clinical signs are not kept alive and euthanized immediately, using carbon dioxide (CO2).

Mice were housed in an accredited facility of the Association for Assessment and Accreditation of Laboratory Animal Care International (AAALAC). This study was carried in strict accordance with the National Institute of Health guidelines for the Care and Use of Laboratory Animals. All procedures in the protocol were approved by the Animal Care and Use Committee (ACUC) of St. Jude Children’s Research Hospital (Animal Assurance Number: A3077-01).

Histology, immunohistochemistry and terminal deoxynucleotidyl transferase dUTP nick end labeling assay

At E14.5 and E15.5, pregnant females received intraperitoneal injections of 5-bromo-2'-deoxyuridine (BrdU at 5 mg/mL and 20 μl/g of mouse), 2 hours before euthanasia. Embryo heads were harvested, fixed overnight in 4% paraformaldehyde (PFA) in phosphate buffered saline (PBS) at 4°C, soaked in 30% sucrose in PBS until the tissues sank to the bottom of the tube, and embedded in Optimal Cutting Temperature (OCT) compound. Frozen 12 μm sections were collected on Fisherbrand superfrost plus slides using a cryostat. Slides were stained with Hematoxylin and Eosin (H&E), with antibodies to Pax6 (PRB-278P; 1/500 dilution; Covance, Emeryville, CA, USA) or BrdU (sc-32323; 1/1000 dilution; Santa Cruz, Santa Cruz, USA), as previously described [33]. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) was performed using the ApoTag Kit (EMD Millipore) following manufacturer’s instructions.

Fluorescence in situ hybridization

Mouse embryo fibroblasts (MEF) were isolated from E14.5 Nestin-Cre embryos, as previously described [34]. Fluorescence In Situ hybridization (FISH) was performed as previously described [35]. In brief, metaphases were hybridized with a biotin labeled chromosome 12 BAC probe (RP23-456E21; CHORI, Oakland CA, USA) and a digoxigenin labeled probe containing the rat Nestin promoter. Probes were visualized by incubation with Texas red avidin and fluorescein labeled anti-digoxigenin.

Quantitative-reverse transcriptase-polymerase chain reaction

Purification of GNPs from P7 cerebella, RNA extraction from dissected embryonic and P7 whole cerebella or from purified GNPs, and Quantitative-Reverse Transcriptase-Polymerase Chain Reaction (Q-RT-PCR) for miR-19a and miR-106b were performed, as previously described [26,33].

Statistics

Statistical significance was determined using GraphPad Prism software (version 5.0). Data were shown as mean +/- s.e.m. P-values <0.05 were used as significance threshold from unpaired two-tailed Student’s t test. For the survival curves, p-values were determined with a log-rank (Mantel Cox) test.

Results

Conditional deletion of Dicer in Nestin-positive progenitor cells

To assess the role of mature microRNAs in brain development, we crossed conditional Dicer-floxed mice with the Nestin-Cre transgenic line and generated Dicerfloxed/floxed; Nestin-Cre+ mice (referred as Dicer cKO). We found that the Nestin-Cre transgene was located on chromosome 12 (band D1) and genetically linked to Dicer (band E-F1) by ~25 megabases (Fig 1A and 1B). Thus, the generation of Dicer cKO animals required that the Nestin-Cre transgene and one floxed allele of Dicer be carried by the same chromosome which necessitated a complex breeding strategy (Fig 1C). Dicer cKO mice died at birth (15/15) but were alive at E18.5. When compared to control littermates (Fig 1D, 1F and 1H), cerebella from E18.5 Dicer cKO embryos had a reduced size and lacked foliations (Fig 1E, 1G and 1I), a thinner EGL visualized with an antibody to Pax6 (Fig 1I) and the rRL devoid of Pax6-expressing progenitor neurons (Fig 1I, white arrows). In contrast, the cerebellum of Dicerfloxed/+; Nestin-Cre+ mice appeared normal (data not shown).

thumbnail
Fig 1. Mice lacking Dicer in Nestin-Cre positive cells display abnormal cerebella.

(A) Chromosome spread from MEFs isolated from a Nestin-Cre embryo were hybridized with a biotin labeled chromosome 12 probe (red dots indicated by red arrows) and a digoxigenin labeled probe containing the rat Nestin promoter (green dots indicated by green arrow). The chromosomes were then stained with DAPI. A representative metaphase spread is shown. (B) Mouse chromosome 12 idiogram showing the location of chromosome 12 probe, Dicer and Nestin-Cre genes. (C) Dicerfloxed/+; Nestin-Cre+ were crossed with C57BL/6 mice to select for Dicerfloxed/+; Nestin-Cre+ in which Dicer and Nestin-Cre were located on the same chromosome and subsequently crossed with Dicerfloxed/floxed; Nestin-Cre- to obtain Dicerfloxed/floxed; Nestin-Cre+ (referred as Dicer cKO) and Dicerfloxed/+; Nestin-Cre- (referred as control). (fl) floxed allele for Dicer. (D, E) Representative mid-sagittal and (F-I) para-sagittal sections through the E18.5 developing cerebellum of control (D, F, H) and Dicer cKO (E, G, I) mice. Tissues stained with H&E (D-G) or a Pax6 antibody (H, I, red). Arrows indicate the rostral rhombic lip. (n = 2 independent embryos; scale bars = 100μm).

https://doi.org/10.1371/journal.pone.0129642.g001

To identify the cause of abnormal cerebellar development, we analyzed embryos at earlier time points. At E14.5, the rRL and the nascent EGL of the Dicer cKO mice appeared similar to control littermates (Fig 2A and 2B). Pax6 positive cells (Pax 6+) were similarly present in the exterior face of the rRL and in the nascent EGL of both Dicer cKO and control mice (Fig 2C and 2D). In contrast, at E15.5, the nascent EGL of the Dicer cKO mice were smaller than in control animals (Fig 2E and 2F), and the exterior face of the rRL appeared thin (Fig 2G and 2H, white arrows). TUNEL staining revealed few apoptotic cells at E14.5 (Fig 2I, 2J and 2Q). In contrast, massive apoptosis was observed in the rRL but not in the EGL, at E15.5 (Fig 2K, 2L and 2Q). In Dicer cKO mice at E15.5 by in vivo BrdU labeling we observed decreased proliferation of the Pax6+ cells in the rRL and EGL (Fig 2P and 2R). Because late-born GNPs contribute to the formation of the posterior EGL [36], the loss of Pax6+ cells in Dicer-deficient posterior cerebella at E18.5 could be explained by the apoptosis of cells in the rRL and their reduced proliferation in the rRL and EGL at E15.5. In addition to the rRL, the Nestin-cre transgene is widely expressed in proliferative zones throughout the central nervous system (CNS). Consistent with this expression pattern, massive apoptosis was observed in the cerebellar ventricular zone (VZ) and caudal rhombic lip (cRL) (Fig 2L and 2Q).

thumbnail
Fig 2. Conditional loss of Dicer induces apoptosis of neural progenitors from the rostral rhombic lip.

Representative sagittal sections through the developing cerebellum of control littermates and Dicer cKO mice at E14.5 (A-D, I, J) and E15.5 (E-H, K, L) stained with H&E (A, B, E, F), an antibody to Pax6 (C, D, G, H, red), TUNEL (I-L green), and a BrdU antibody (M-P, green). Nuclei were counterstained with propidium iodide (I-L, red). Black and white arrows indicate the rRL. E14.5 n = 2, E15.5 n = 3, independent embryos. Black and white scale bars = 100μm. (Q) Number of TUNEL+ cells were counted on a 1.44mm2 surface within the ventricular zone (VZ) and the rostral rhombic lip (rRL) from 2 independent brain sections of one embryo per genotype. (R) Percentage of Pax6+, BrdU+ cells in the rRL and the external granule layer (EGL) of n = 3 control and n = 3 Dicer cKO mice. (**) p-value = 0.0073.

https://doi.org/10.1371/journal.pone.0129642.g002

Apoptosis was already detected in the neocortex of Dicer cKO embryos from E14.5 onward (Fig 3A3D and 3G), as previously reported [10,12] and in the VZ of the colliculus (data not shown). The VZ of the neocortex generates neurons that radially migrate towards the surface of the brain to their final position with a specific address dependent on their birth date resulting in a cortex with characteristic six neuronal layers (Fig 3E). In the Dicer cKO mice, at E18.5, the cortex was less laminated with a thin VZ (Fig 3F).

thumbnail
Fig 3. Massive apoptosis in the cerebral cortex of the Dicer cKO embryos.

Representative TUNEL staining (green) was performed on sagittal sections of the cortex (A-D) from a control (A, C) and Dicer cKO (B, D) mouse at E14.5 (A, B) and E15.5 (C, D). Nuclei were counterstained with propidium iodide (red). Representative sagittal sections through the cortex of an E18.5 control (E) and Dicer cKO (F) mouse were stained with H&E. Vertical black bars indicate the ventricular zone (VZ). n = 2 independent embryos. Black and white scale bars = 100μm. (G) TUNEL+ cells were quantified in the VZ from 2 independent brain sections of one embryo per genotype.

https://doi.org/10.1371/journal.pone.0129642.g003

Apoptosis can proceed by a Trp53-dependent pathway, particularly in the case of induced genome instability, or by Trp53-independent pathways [37, 38]. To determine if the cell death resulting from Dicer loss was a result of DNA damage, we asked if the apoptosis was dependent on Trp53 function. Loss of Trp53 did not rescue lethality, nor the phenotypes observed in the developing brain of the Dicer cKO mice (i.e. apoptosis, abnormal cerebellum and less laminated cortex) (Fig 4A4M), suggesting that apoptosis and other phenotypes did not result from DNA damage.

thumbnail
Fig 4. Deletion of Trp53 is unable to rescue the phenotypes observed in Dicer cKO mice.

Representative sagittal sections through the developing cerebellum (A-C, G-L) and cortex (D-F) of Dicer cKO (A, D, G, J), Dicer cKO; Trp53 KO (B, E, H, K, and Trp53 KO (C, F, I, L) mice at E18.5 (A-F) and E15.5 (G-L) were stained with H&E (A-I) and by TUNEL (J-L). Nuclei were counterstained with propidium iodide (red) (J-L). Black and white arrows indicate the rostral rhombic lip. Vertical black bars indicate the ventricular zone (VZ). (n = 2 independent E18.5 or 3 E15.5 embryos. Scale bars = 100μm). (M) TUNEL+ cells were counted in the VZ and rRL from 2 independent brain sections of 3 embryos per genotypes. (***) p-values = 0.0002; (**) p-values = 0.0014 and ns (non-significant)

https://doi.org/10.1371/journal.pone.0129642.g004

MicroRNAs are progressively deleted in the cerebellum of Dicer cKO embryos

Cre recombinase activity under the control of the Nestin promoter is initiated between E9.5 and E10.5 [39]. However, we observed massive apoptosis at E15.5 in the rRL and the VZ of the developing cerebella in Dicer cKO mice, suggesting that complete loss of Dicer and therefore of mature microRNAs may have happened progressively from E9.5 to E15.5. We confirmed that the two Dicer alleles were deleted in the cerebellum at E14.5 (Fig 5A). We previously reported that the miR-17~92 cluster family is expressed in the developing cerebellum [29]. To evaluate how the loss of Dicer affects the expression of mature microRNAs, we determined the expression of miR-19a (from the miR-17~92 cluster) and miR-106b (from the miR-106b~25 cluster) in the developing cerebella of control and Dicer cKO embryos at E14.5, E16.5 and E18.5 by Q-RT-PCR (Fig 5B and 5C, lanes 1–6). While the levels of miR-19a and miR-106b were not statistically different at E14.5 (Fig 5B and 5C, lane 1 versus lane 4), their levels were significantly reduced at E16.5 (Fig 5B and 5C, lane 2 versus lane 5, p = 0.0386 and 0.0001, respectively) and at E18.5 (Fig 5B and 5C, lane 3 versus lane 6, p = 0.0005 and 0.0005, respectively). Therefore deletion of Dicer resulted in a delayed decrease of miR-19a and miR-106b microRNAs with significant loss of those microRNAs at E16.5.

thumbnail
Fig 5. Delayed loss of microRNAs in Dicer cKO embryos.

(A) PCR for Dicer and Nestin-Cre was performed on DNA extracted from tail (lane 1) or total cerebella (lanes 2–11) of Dicer cKO mice at E14.5 (lanes 1–5), E16.5 (lanes 6–8) and E18.5 (lanes 9–11); Dicer alleles: (fl) floxed, (Δ) deleted. Relative levels of mature microRNAs miR-19a (B) and miR-106b (C) were determined by Q-RT-PCR on total RNA extracted from total cerebella of control (lanes 1–3) and Dicer cKO (lanes 4–6) mice at E14.5 (lanes 1, 4), E16.5 (lanes 2, 5) and E18.5 (lanes 3, 6). (*) p-values <0.05, (***, ###) p-values <0.001. n = 3 independent samples. (*) p-values were calculated by comparing lane 2 to lane 5 and (#) lane 3 to lane 6.

https://doi.org/10.1371/journal.pone.0129642.g005

Dicer restrains medulloblastoma development

Dicer was previously reported to be haploinsufficient for tumor suppression in several mouse and human cancers [19,20]. SHH medulloblastoma arise from GNPs with aberrant SHH signaling upon deletion of one copy of the SHH receptor Patched (Ptch1+/-) alone or with one or two copies of Cdkn2c [33]. To determine if Dicer was haploinsufficient for SHH medulloblastoma development, Dicerfloxed/+; Nestin-Cre+ animals were bred to the Ptch1+/-; Cdkn2c-/- mice. Loss of one copy of Dicer induced SHH medulloblastoma with a similar time of onset and penetrance in Dicerfloxed/+; Nestin-Cre+; Ptch1+/-; Cdkn2c+/- compared to Dicer+/+; Ptch1+/-; Cdkn2c+/- mice, 76.5% (39/51) versus 62% (31/50), respectively (Fig 6A). In contrast, Dicerfloxed/+; Nestin-Cre+; Ptch1+/-; Cdkn2c+/- mice died significantly faster (median survival, 144 days) than Dicer+/+; Ptch1+/-; Cdkn2c+/- mice (198.5 days) (p = 0.0247) (Fig 6A). We confirmed by PCR that the floxed allele but not the wild type allele of Dicer was deleted (Fig 6B, lanes 1–4). Analysis of tumors pathology from the two genotypes did not reveal differences and confirmed that they were medulloblastoma of the SHH subgroup with classic morphology of round cells (Fig 6C6F). These results suggest that Dicer restrains SHH medulloblastoma development and is haploinsufficient for tumor suppression.

thumbnail
Fig 6. Dicer restrains medulloblastoma development.

(A) Survival curves for Ptch1+/-; Cdkn2c+/- mice with different Dicer and Nestin-Cre genotypes. Dicerfloxed/+; Nestin-Cre+; (green line, n = 51) and Dicer+/+ (red line, n = 50). (B) PCR for Dicer and Nestin-Cre was performed on DNA extracted from tail (T1) of a Dicerfloxed/floxed; Nestin-Cre-; Ptch1+/-; Cdkn2c+/- mouse, tail (T2) and two independent pieces of the same medulloblastoma (MBa and MBb) from a Dicerfloxed/+; Nestin-Cre+; Ptch1+/-; Cdkn2c+/- mouse. Dicer alleles: (fl) floxed, (+) wild type, (Δ) deleted. (C-F) Representative H&E staining of medulloblastoma in (C) Dicerfloxed/+; Nestin-Cre+; Ptch1+/-; Cdkn2c+/- and (E) Dicer+/+; Ptch1+/-; Cdkn2c+/- mice. Scale bars = 500μm (C and E) and 50μm (D and F).

https://doi.org/10.1371/journal.pone.0129642.g006

Discussion

We here report the role of Dicer and mature miRNAs during cerebellar development and SHH medulloblastoma genesis by conditional deletion of Dicer (Dicer cKO) in Nestin-positive neural progenitors. We found that conditional inactivation of Dicer triggered extensive Trp53-independent apoptosis in all proliferative zones throughout the developing CNS and decreased proliferation of GNPs in the developing cerebellum. Loss of one copy of Dicer accelerated medulloblastoma formation in Ptch1+/-, Cdkn2c+/- animals. Dicer was shown by several groups to be required for proper development of many neural cell populations in the developing CNS [814,18]. Conditional loss of Dicer in the CNS was previously found to induce apoptosis using the same [10,12] or different [8,9,11,13,17,18,40] Cre lines, and to affect cortical development [810,12]. While using the same Nestin-Cre and Dicer-floxed lines, s, the group of Kawasa-Koga did not find obvious defects cerebellar anomalies in their Dicer cKO mice [10]. They show that while miRNAs are still detected at E15.5 in the cortex of their animals, they are absent at E18.5 correlating with apoptosis. Kawasa-Koga and colleagues remarked that Nestin-Cre; Dicer mice produced very few Dicer conditional knockout mice embyos, 6.8% at E18.5 and that no surviving newborns were detected. This is consistent with our data and the finding that Dicer and Nestin-Cre are genetically linked on mouse chromosome 12. In the Discussion of their paper, Kawase-Koga and colleagues did not find obvious defects in cerebellar development during embryonic stages and yet, they obtained very few and no live embryos while detecting defects in the cortex of Nestin-Cre; Dicer mice. These differences in phenotypes between groups may potentially be due to the genetic background. Using the same Nestin-Cre but different Dicer-floxed mice, McLoughlin and collaborators also describe cortical defects in Dicer cKO mice, but did not report on the status of the mice cerebella [12]. McLoughlin and colleagues used Dicer-floxed animals generated by Harfe and colleagues. In their manuscript, they only described the effect of Dicer deletion in the cortex but not in the cerebellum and found increased apoptosis in cortical regions at E15.5, which is consistent with our data. They also detected low numbers of Dicer conditional knockout mice embryos and no surviving embryos. Kawase-Koga and McLoughlin focused on cortical development while we assessed the role of Dicer in the embryonic cerebellum. To increase the percentage of embryos with the proper genotype, we backcrossed our mice into C57BL/6 background (Fig 1C) that changed the genetic background of our cohort, compared to that of Kawase-Koga and colleagues. This may explain why Kawase-Koga and colleagues do not see apoptosis at E15.5 in the cortices of Nestin-Cre+; Dicerfloxedl/floxed mice; even though they show, by immunoblotting, that the levels of Dicer protein are greatly reduced at E15.5. They also note that Dicer deletion occurs at different times depending on the localization.

Despite these few differences, our results are consistent with these reports since conditional deletion of Dicer in Nestin-positive cells induced substantial apoptosis in all proliferative zones of the CNS.

In Dicer cKO mice, loss of neural progenitors in the rRL led to a diminished pool of GNPs which resulted in an abnormally developed cerebellum with a short and thin EGL, absence of folia and an aberrant residual rRL at E18.5. This was associated with apoptosis in the rRL and reduced proliferation of GNPs in the developing EGL. Apoptosis was also observed in the ventricular zone of the developing cerebellum. These results suggest that lack of Dicer induced apoptosis in stem / progenitor neural cells, before they commit to their final lineages and controlled the proliferation of GNPs in the developing EGL

Although Nestin is expressed as early as E9.5–10.5 in neural progenitors [39], the decrease in microRNAs levels in the developing cerebella of the Dicer cKO mice was delayed, consistent with the relatively long half-life of miRNAs [41]. At E14.5, the cerebella of Dicer cKO and control littermates were normal and the levels of miRs were not significantly reduced. However, at E15.5, massive apoptosis was observed, suggesting that residual levels of microRNAs were insufficient to maintain the proliferation of neural progenitors. Overall, our results suggest that processing of microRNAs by Dicer was required for proper cerebellar development during embryogenesis. Alternatively, the effects observed might be due to miR-independent cell survival function of Dicer, as previously reported [42]. Thus, our study is the first analysis of the role of Dicer during cerebellar development. We consider that our data and that from others illustrate a critical role for this protein, in the regulation of miRNAs during development of the cerebellum.

We found that Dicer restricts SHH medulloblastoma development in Ptch1+/- mice. Our results are consistent with those reported in two mouse models of retinoblastoma and lung cancers [19,20] but not in Eμ-Myc B cell lymphoma [21]. We previously showed that miR-17~92 expression is absolutely required for SHH medulloblastoma initiation [29] and that its silencing inhibits medulloblastoma progression [43]. This implies that complete loss of Dicer is selected against medulloblastoma formation and progression. Here we show that loss of one copy of Dicer in a SHH medulloblastoma mouse model accelerated tumor formation, suggesting that decreased levels of some mature tumor suppressor miRNAs might promote tumorigenesis. We will unable to correlate the phenotypes of Dicer cKO in embryos with the ones we observed with the mir17~92 cluster family because the effects of the loss of the mir17~92 cluster family are only revealed in the cerebellum after birth [29]. The Dicer cKO mice instead die at birth. We previously published that the miR-17~92 cluster family was required for proliferation of GNPs during postnatal cerebellar and medulloblastoma development [29,43]. In the present study, we used two of the microRNAs encoded by the miR-17~92 cluster family (miR-19a and miR106b) to determine the level of microRNA biogenesis. In previous studies, we determined that these two miRNAs were expressed during embryonic cerebellar development [29]. Clearly other microRNAs besides those encoded by the miR-17~92 cluster family are expressed during cerebellar development. Therefore, the apoptosis observed at E15.5 Dicer cKO embryos is most likely due, only in part, to the loss of expression of microRNAs encoded by the miR-17~92 cluster family.

In summary, while Dicer is essential for proper cerebellar development and homeostasis, it restrains SHH medulloblastoma development suggesting that it might be haploinsufficient for medulloblastoma suppression.

Acknowledgments

We are indebted to Dr. Gregory Hannon for graciously providing the conditional Dicer animals and to Dr. David Adler for the idiogram of chromosome 12 (http://www.pathology.washington.edu/research/cytopages/idiograms/mouse/). We thank Shelly Wilkerson, Sarah Robinson and Dana Farmer for mouse genotyping, immunohistochemistry experiments and mouse colony management. We thank Mark and Virginia Valentine for FISH analysis, Brent Orr for pathology analysis. We also thank Drs. Charles J. Sherr, David Solecki, Mark Hatley, Brandon Wainwright and all members of the laboratory for helpful discussions and suggestions during the course of these experiments.

Author Contributions

Conceived and designed the experiments: FZ MFR. Performed the experiments: FZ Y. Lee DK OA LBM Y. Li. Analyzed the data: FZ Y. Lee DK OA Y. Li PJM MFR. Wrote the paper: FZ PJM MFR.

References

  1. 1. Carmell MA, Hannon GJ. RNase III enzymes and the initiation of gene silencing. Nat Struct Mol Biol. 2004; 11: 214–218. pmid:14983173
  2. 2. Kim VN. MicroRNA biogenesis: coordinated cropping and dicing. Nat Rev Mol Cell Biol. 2005; 6: 376–385. pmid:15852042
  3. 3. Bernstein E, Kim SY, Carmell MA, Murchison EP, Alcorn H, Li MZ, et al. Dicer is essential for mouse development. Nat Genet. 2003; 35: 215–217. pmid:14528307
  4. 4. Soukup GA, Fritzsch B, Pierce ML, Weston MD, Jahan I, McManus MT, et al. Residual microRNA expression dictates the extent of inner ear development in conditional Dicer knockout mice. Dev Biol. 2009; 328: 328–341. pmid:19389351
  5. 5. Friedman LM, Dror AA, Mor E, Tenne T, Toren G, Satoh T, et al. MicroRNAs are essential for development and function of inner ear hair cells in vertebrates. Proc Natl Acad Sci U S A. 2009; 106: 7915–7920. pmid:19416898
  6. 6. Damiani D, Alexander JJ, O'Rourke JR, McManus M, Jadhav AP, Cepko CL, et al. Dicer inactivation leads to progressive functional and structural degeneration of the mouse retina. J Neurosci. 2008; 28: 4878–4887. pmid:18463241
  7. 7. Georgi SA, Reh TA. Dicer is required for the transition from early to late progenitor state in the developing mouse retina. J Neurosci. 2010; 30: 4048–4061. pmid:20237275
  8. 8. Davis TH, Cuellar TL, Koch SM, Barker AJ, Harfe BD, McManus MT, et al. Conditional loss of Dicer disrupts cellular and tissue morphogenesis in the cortex and hippocampus. J Neurosci. 2008; 28: 4322–4330. pmid:18434510
  9. 9. 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: 3911–3921. pmid:18997113
  10. 10. Kawase-Koga Y, Otaegi G, Sun T. Different timings of Dicer deletion affect neurogenesis and gliogenesis in the developing mouse central nervous system. Dev Dyn. 2009; 238: 2800–2812. pmid:19806666
  11. 11. Li Q, Bian S, Hong J, Kawase-Koga Y, Zhu E, Zheng Y, et al. Timing specific requirement of microRNA function is essential for embryonic and postnatal hippocampal development. PLoS One. 2011; 6: e26000. pmid:21991391
  12. 12. McLoughlin HS, Fineberg SK, Ghosh LL, Tecedor L, Davidson BL. Dicer is required for proliferation, viability, migration and differentiation in corticoneurogenesis. Neuroscience. 2012; 223: 285–295. pmid:22898830
  13. 13. Schaefer A, O'Carroll D, Tan CL, Hillman D, Sugimori M, Llinas R, et al. Cerebellar neurodegeneration in the absence of microRNAs. J Exp Med. 2007; 204: 1553–1558. pmid:17606634
  14. 14. Cuellar TL, Davis TH, Nelson PT, Loeb GB, Harfe BD, Ullian E, et al. Dicer loss in striatal neurons produces behavioral and neuroanatomical phenotypes in the absence of neurodegeneration. Proc Natl Acad Sci U S A. 2008; 105: 5614–5619. pmid:18385371
  15. 15. Pang X, Hogan EM, Casserly A, Gao G, Gardner PD, Tapper AR. Dicer expression is essential for adult midbrain dopaminergic neuron maintenance and survival. Mol Cell Neurosci. 2014; 58: 22–28. pmid:24184162
  16. 16. Bremer J, O'Connor T, Tiberi C, Rehrauer H, Weis J, Aguzzi A. Ablation of Dicer from murine Schwann cells increases their proliferation while blocking myelination. PLoS One. 2010; 5: e12450. pmid:20805985
  17. 17. Tao J, Wu H, Lin Q, Wei W, Lu XH, Cantle JP, et al. Deletion of astroglial Dicer causes non-cell-autonomous neuronal dysfunction and degeneration. J Neurosci. 2011; 31: 8306–8319. pmid:21632951
  18. 18. Nowakowski TJ, Mysiak KS, Pratt T, Price DJ. Functional dicer is necessary for appropriate specification of radial glia during early development of mouse telencephalon. PLoS One. 2011; 6: e23013. pmid:21826226
  19. 19. Kumar MS, Pester RE, Chen CY, Lane K, Chin C, Lu J, et al. Dicer1 functions as a haploinsufficient tumor suppressor. Genes Dev. 2009; 23: 2700–2704. pmid:19903759
  20. 20. Lambertz I, Nittner D, Mestdagh P, Denecker G, Vandesompele J, Dyer MA, et al. Monoallelic but not biallelic loss of Dicer1 promotes tumorigenesis in vivo. Cell Death Differ. 2010; 17: 633–641. pmid:20019750
  21. 21. Arrate MP, Vincent T, Odvody J, Kar R, Jones SN, Eischen CM. MicroRNA biogenesis is required for Myc-induced B-cell lymphoma development and survival. Cancer Res. 2010; 70: 6083–6092. pmid:20587524
  22. 22. Ravi A, Gurtan AM, Kumar MS, Bhutkar A, Chin C, Lu V, et al. Proliferation and tumorigenesis of a murine sarcoma cell line in the absence of DICER1. Cancer Cell. 2012; 21: 848–855. pmid:22698408
  23. 23. Slade I, Bacchelli C, Davies H, Murray A, Abbaszadeh F, Hanks S, et al. DICER1 syndrome: clarifying the diagnosis, clinical features and management implications of a pleiotropic tumour predisposition syndrome. J Med Genet. 2011; 48: 273–278. pmid:21266384
  24. 24. Foulkes WD, Priest JR, Duchaine TF. DICER1: mutations, microRNAs and mechanisms. Nat Rev Cancer. 2014; 14: 662–672. pmid:25176334
  25. 25. Hatten ME, Roussel MF. Development and cancer of the cerebellum. Trends Neurosci. 2011; 34: 134–142. pmid:21315459
  26. 26. Uziel T, Karginov FV, Xie S, Parker JS, Wang YD, Gajjar A, et al. The miR-17~92 cluster collaborates with the Sonic Hedgehog pathway in medulloblastoma. Proc Natl Acad Sci U S A. 2009; 106: 2812–2817. pmid:19196975
  27. 27. Northcott PA, Fernandez LA, Hagan JP, Ellison DW, Grajkowska W, Gillespie Y, et al. The miR-17/92 polycistron is up-regulated in sonic hedgehog-driven medulloblastomas and induced by N-myc in sonic hedgehog-treated cerebellar neural precursors. Cancer Res. 2009; 69: 3249–3255. pmid:19351822
  28. 28. Shalaby T, Fiaschetti G, Baumgartner M, Grotzer MA. MicroRNA signatures as biomarkers and therapeutic target for CNS embryonal tumors: the pros and the cons. Int J Mol Sci. 2014; 15: 21554–21586. pmid:25421247
  29. 29. Zindy F, Kawauchi D, Lee Y, Ayrault O, Ben Merzoug L, McKinnon PJ, et al. Role of the miR-17 approximately 92 cluster family in cerebellar and medulloblastoma development. Biol Open. 2014; 3: 597–605. pmid:24928431
  30. 30. Murchison EP, Partridge JF, Tam OH, Cheloufi S, Hannon GJ. Characterization of Dicer-deficient murine embryonic stem cells. Proc Natl Acad Sci U S A. 2005; 102: 12135–12140. pmid:16099834
  31. 31. Jacks T, Remington L, Williams BO, Schmitt EM, Halachmi S, Bronson RT, et al. Tumor spectrum analysis in p53-mutant mice. Curr Biol. 1994; 4: 1–7. pmid:7922305
  32. 32. Tronche F, Kellendonk C, Kretz O, Gass P, Anlag K, Orban PC, et al. Disruption of the glucocorticoid receptor gene in the nervous system results in reduced anxiety. Nat Genet. 1999; 23: 99–103. pmid:10471508
  33. 33. Uziel T, Zindy F, Xie S, Lee Y, Forget A, Magdaleno S, et al. The tumor suppressors Ink4c and p53 collaborate independently with Patched to suppress medulloblastoma formation. Genes Dev. 2005; 19: 2656–2667. pmid:16260494
  34. 34. Zindy F, Quelle DE, Roussel MF, Sherr CJ. Expression of the p16INK4a tumor suppressor versus other INK4 family members during mouse development and aging. Oncogene. 1997; 15: 203–211. pmid:9244355
  35. 35. Roberts KG, Li Y, Payne-Turner D, Harvey RC, Yang YL, Pei D, et al. Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia. N Engl J Med. 2014; 371: 1005–1015. pmid:25207766
  36. 36. Machold R, Fishell G. Math1 is expressed in temporally discrete pools of cerebellar rhombic-lip neural progenitors. Neuron. 2005; 48: 17–24. pmid:16202705
  37. 37. Clarke AR, Purdie CA, Harrison DJ, Morris RG, Bird CC, Hooper ML, et al. Thymocyte apoptosis induced by p53-dependent and independent pathways. Nature. 1993; 362: 849–852. pmid:8479523
  38. 38. McKinnon PJ. Maintaining genome stability in the nervous system. Nat Neurosci. 2013; 16: 1523–1529. pmid:24165679
  39. 39. Graus-Porta D, Blaess S, Senften M, Littlewood-Evans A, Damsky C, Huang Z, et al. Beta1-class integrins regulate the development of laminae and folia in the cerebral and cerebellar cortex. Neuron. 2001; 31: 367–379. pmid:11516395
  40. 40. Huang T, Liu Y, Huang M, Zhao X, Cheng L. Wnt1-cre-mediated conditional loss of Dicer results in malformation of the midbrain and cerebellum and failure of neural crest and dopaminergic differentiation in mice. J Mol Cell Biol. 2010; 2: 152–163. pmid:20457670
  41. 41. Gantier MP, McCoy CE, Rusinova I, Saulep D, Wang D, Xu D, et al. Analysis of microRNA turnover in mammalian cells following Dicer1 ablation. Nucleic Acids Res. 2011; 39: 5692–5703. pmid:21447562
  42. 42. Kaneko H, Dridi S, Tarallo V, Gelfand BD, Fowler BJ, Cho WG, et al. DICER1 deficit induces Alu RNA toxicity in age-related macular degeneration. Nature. 2011; 471: 325–330. pmid:21297615
  43. 43. Murphy BL, Obad S, Bihannic L, Ayrault O, Zindy F, Kauppinen S, et al. Silencing of the miR-17~92 cluster family inhibits medulloblastoma progression. Cancer Res. 2013; 73: 7068–7078. pmid:24145352