Medulloblastoma, originating in the cerebellum, is the most common malignant brain tumor in children. Medulloblastoma consists of four major groups where constitutive activation of the Sonic Hedgehog (SHH) signaling pathway is a hallmark of one group. Mouse and human SHH medulloblastomas exhibit increased expression of microRNAs encoded by the miR-17∼92 and miR-106b∼25 clusters compared with granule progenitors and postmitotic granule neurons. Here, we assessed the therapeutic potential of 8-mer seed-targeting locked nucleic acid (LNA)-modified anti-miR oligonucleotides, termed tiny LNAs, that inhibit microRNA seed families expressed by miR-17∼92 and miR-106b∼25 in two mouse models of SHH medulloblastomas. We found that tumor cells (medulloblastoma cells) passively took up 8-mer LNA-anti-miRs and specifically inhibited targeted microRNA seed-sharing family members. Inhibition of miR-17 and miR-19a seed families by anti-miR-17 and anti-miR-19, respectively, resulted in diminished tumor cell proliferation in vitro. Treatment of mice with systemic delivery of anti-miR-17 and anti-miR-19 reduced tumor growth in flank and brain allografts in vivo and prolonged the survival of mice with intracranial transplants, suggesting that inhibition of the miR-17∼92 cluster family by 8-mer LNA-anti-miRs might be considered for the treatment of SHH medulloblastomas. Cancer Res; 73(23); 7068–78. ©2013 AACR.

Medulloblastoma, a cerebellar tumor, is the most common malignant pediatric brain cancer (1). Molecular profiling classifies human medulloblastomas into four major subgroups with distinct mRNA and miRNA signatures: Sonic Hedgehog (SHH), WNT, group 3 (G3), and group 4 (G4; ref. 2). As current therapies induce adverse effects that compromise patients' quality of life (3, 4), novel therapeutic options have become a necessity. Small-molecule inhibitors of Smoothened (SMO) are effective at eliminating murine (5) and human (6) SHH medulloblastomas. However, treatment of young mice with the SHH antagonist induces long-bone defects (7). In the clinic, one patient with significant remission after treatment with GDC-0449 developed resistance due to SMO mutation (8). These recent results suggested that other approaches are necessary to treat SHH medulloblastomas.

MicroRNAs (miRNA) are endogenous noncoding RNAs of approximately 22 nucleotides, which play key roles in numerous biologic processes, including cancer (9). They bind to partially complementary sequences within the 3′-untranslated regions (UTR) of target mRNAs inducing translational repression or mRNA degradation (10). The miR-17∼92 cluster belongs to a family of 3 clusters encoded on different chromosomes (11). They include miR-17∼92, also called OncomiR-1, which encodes 6 miRNAs (miR-17, miR-18a, miR-19a, miR-20a, miR-19b-1, and miR-92-1), miR-106b∼25, which encodes 3 miRNAs (miR-106b, miR-93m and miR-25), and miR-106a∼363, which encodes 6 miRNAs (miR-106a, miR-18b, miR-20b, miR-19b-2, miR-92-2, and miR-363). Individual miRNAs are classified into 4 families based on sequence similarities within their 6-nucleotide seed sequence (Fig. 1A; refs. 11, 12). The oncogenic potential of miR-17∼92 was discovered in B-cell lymphomas (13), with the miR-19a/b family responsible for tumorigenicity (14, 15).

Figure 1.

MicroRNAs encoded by the miR-17∼92 cluster in medulloblastoma. A, miRNAs encoded by the three clusters divided into four families according to their shared seed sequences, denoted by color coding. miR-17 and miR-19 seed families were targeted by 8-mer LNA-anti-miRs: complementary sequences to miR-17 and miR-19a are shown in red and blue, respectively, adapted from the work of Ventura and colleagues (11). B, relative miRNA levels were quantified from RNAs extracted from medulloblastomas that developed in [Ptch1+/−;Cdkn2c−/−] mice (n = 6), spontaneous medulloblastomas arisen from [Ptch1+/−;Trp53−/−] mice (n = 4), and whole cerebellum of 1-month-old mice (1 mo Cb; n = 3). Error bars, SD. *, **, and ***, P ≤ 0.05, P ≤ 0.01, and P ≤ 0.001 compared with 1-mo Cb.

Figure 1.

MicroRNAs encoded by the miR-17∼92 cluster in medulloblastoma. A, miRNAs encoded by the three clusters divided into four families according to their shared seed sequences, denoted by color coding. miR-17 and miR-19 seed families were targeted by 8-mer LNA-anti-miRs: complementary sequences to miR-17 and miR-19a are shown in red and blue, respectively, adapted from the work of Ventura and colleagues (11). B, relative miRNA levels were quantified from RNAs extracted from medulloblastomas that developed in [Ptch1+/−;Cdkn2c−/−] mice (n = 6), spontaneous medulloblastomas arisen from [Ptch1+/−;Trp53−/−] mice (n = 4), and whole cerebellum of 1-month-old mice (1 mo Cb; n = 3). Error bars, SD. *, **, and ***, P ≤ 0.05, P ≤ 0.01, and P ≤ 0.001 compared with 1-mo Cb.

Close modal

Several mouse models of SHH medulloblastomas exist with mutation of Smo (16), Ptch1 alone (17), or with loss of Trp53 (18) or the cyclin-dependent kinase–inhibitory proteins, p18Ink4c/Cdkn2c or p27Kip1/Cdkn1b (19, 20). Mouse and human SHH medulloblastomas exhibit high levels of miRNAs encoded by miR-17∼92 and miR-106b∼25 (21, 22) with amplification of miR-17∼92 found in less than 10% of human medulloblastomas (22). Enforced expression of miR-17∼92 increases proliferation of postnatal day 7 (P7) cerebellar granule neuron progenitors (GNP; ref. 22) and collaborates with mutated Ptch1 to induce medulloblastoma in mice (21). We hypothesized that targeting members of the miRNA seed families from miR-17∼92 and miR-106b∼25 could serve as potential therapies for the treatment of SHH medulloblastomas. We recently reported that tiny locked nucleic acids (LNA) inhibit the function of miRNAs, including miRNA seed families (23–25). Here, we investigated the therapeutic potential of 8-mer LNA-anti-miRs in inhibiting miR-17, 20a, 106b, and 93 (anti-miR-17) and miR-19a and 19b-1 (anti-miR-19) in 2 murine SHH medulloblastoma models.

Animal husbandry

[Ptch1+/−;Trp53−/−] and [Ptch1+/−;Cdkn2c−/−] mice develop SHH medulloblastomas. Transplants were conducted in 1- to 2-month-old CD-1 nude (CD-1) mice (Charles River Laboratories). [NestinCre+/−;miR-17∼92floxed/floxed;miR-106b-25−/−] double knockout (DKO) mice were generated by conditionally deleting miR-17∼92 (11) in cells expressing the Cre recombinase driven by the Nestin promoter (26) in an miR-106b∼25--null background (11). Mice were housed in an accredited facility of the Association for Assessment of Laboratory Animal Care in accordance with the NIH guidelines. The Institutional Animal Care and Use Committee of SJCRH approved all procedures in this study.

Purification, culture, and LNA-anti-miR treatment of medulloblastoma cells

Primary medulloblastoma cells from spontaneously arisen SHH medulloblastomas from [Ptch1+/−;Cdkn2c−/−] mice were purified and cultured as previously described (27). Medulloblastoma cells were treated with tiny LNAs targeting the seed region of miR-17, miR-19a, or the 8-mer scrambled LNA control (referred to as anti-miR-17, anti-miR-19, and scrambled LNA). After 48 hours, medulloblastoma cells were harvested with trypsin (Worthington Biochemicals) and ovomucoid (trypsin inhibitor, Worthington Biochemicals) solutions for further analysis. Fluorescence-activated cell sorting (FACS) was used to assess bromodeoxyuridine (BrdUrd) incorporation following a 2-hour pulse of 10 μmol/L BrdUrd (BD Pharmingen, 552598) and the percentage of Annexin V–positive cells. To determine the efficiency of uptake, medulloblastoma cells were treated with 8 μmol/L 6-carboxyfluorescein (FAM) tiny LNAs. Tiny LNA sequences were (5′–3′): anti-miR-17, AGCACTTT; anti-miR-19, ATTTGCAC; scrambled LNA control, TCATACTA, as previously published (23).

Medulloblastoma allografts

Pretreatment with tiny LNAs before transplant.

A total of 2 × 106 medulloblastoma cells were treated with tiny LNAs during a 30-minute preplating step, harvested, resuspended in Matrigel (BD Biosciences), and injected subcutaneously into the flank of CD-1 mice. A subset of mice was sacrificed 14 days after transplant when flank tumors were palpable. Tumors were sectioned and stained for the FAM label using an anti-FAM–conjugated AlexaFluor 594. The presence of tiny LNAs was visualized by confocal microscopy. The remaining mice were imaged weekly using the VEVO-770 High-Resolution 3-Dimension Ultrasound Imaging System, and tumor volumes were measured by tracing tumors within virtual sections of the 3D image stack (VisualSonics). Mice were sacrificed when the size of the tumor was greater than 20% of the total animal body mass.

Intravenous tiny LNA administration after transplant.

Three spontaneous medulloblastomas from [Ptch1+/−;Trp53−/−] mice were maintained by transplantation in the flank of CD-1 mice. Each tumor was purified and infected with lentiviruses encoding a spleen focus-forming virus-LTR driving 2A peptide–linked luciferase and yellow fluorescent protein (YFP; CL20-luc2aYFP). Forty-eight hours later, cells were subjected to FACS and 1 × 106 YFP-positive cells were transplanted into the left flank of recipient animals (day 0). For cranial transplants, 0.5 × 106 YFP-positive cells were injected into the cortices of CD-1 mice, as described previously (21). Bioluminescence imaging of luciferase activity monitored tumor growth twice weekly using a Xenogen IVIS-200 system prior and during LNA treatment. Three milligrams of d-luciferin (Caliper Life Sciences) was administered to each mouse intraperitoneally, 5 to 7 minutes before bioluminescence imaging. Radiance (photons/s/cm2/steridian) was determined within regions of interest (flank or head of mice) using Living Image Software version 4.3.1 (Caliper Life Sciences). Bioluminescence imaging detected luciferase in medulloblastoma cells the day after transplant (day 1) with an intensity ≥ 1.3 × 106 radiance. Mice received intravenous (i.v.) injections via tail vein of saline-formulated tiny LNAs (scrambled, anti-miR-17, or anti-miR-19) or an equal volume of saline, with an initial loading dose of 25 mg/kg (day 2). For flank transplants, maintenance doses of tiny LNAs were administered twice weekly at 10 mg/kg (days 6, 9, 13, and 16). Mice were sacrificed 24 to 72 hours after the last dose of tiny LNAs. For cranial transplants, maintenance doses of 20 mg/kg were given on days 6 and 9. Mice were sacrificed when they became lethargic and displayed neurological signs (e.g., head tilting, head dome, failure to thrive), which occurred 5 to 8 days after transplant for mice treated with saline or scrambled LNA.

Blood–brain barrier integrity

Three mice with intracranial transplantation of 0.5 × 106 YFP-positive medulloblastoma cells from one [Ptch1+/−;Trp53−/−] tumor were injected i.v. with either 2 mL/kg of 2% Evans blue (Sigma-Aldrich, E2129) in 0.9% saline 6 days after transplant or 25 mg/kg FAM-labeled scrambled LNA 2 weeks after transplant. Evans blue is a dye with a high affinity for serum albumin not normally present in the brain (28, 29). Mice that received Evans blue were euthanized by cardiac perfusion with 4% paraformaldehyde 24 hours later. Mice that received tiny LNA injections were perfused after 30 and 60 minutes. Brains were cryoprotected in sucrose, frozen and sectioned for immunohistochemistry.

RNA extraction, quantitative real-time PCR

Total RNA from spontaneous medulloblastomas, purified tumor cells and flank allografts was extracted, reverse transcribed, and quantitative real-time PCR (qRT-PCR) for miRNAs was conducted, as described previously [ref. 21; ABI, catalogue number 4427975 and assay ID numbers = 001093 002308 (RNU6B), (hsa-miR-17), 002422 (hsa-miR-18a), 000580 (hsa-miR-20a), 000395 (hsa-miR-19a), 000396 (hsa-miR-19b), 000430 (hsa-miR-92), 000442 (hsa-miR-106b), 001090 (mmu-miR-93)]. Relative levels of microRNAs were quantified in triplicate. Ct values were normalized within the same RNA sample to the small nuclear RNA RNU6B and to P6 GNPs (relative value = 1).

Plasmid construction, cell culture, and 3′-UTR luciferase assay

The luciferase assay was conducted using the pmirGLO dual-luciferase miRNA target expression vector (pmirGLO; Promega). Oligonucleotides containing predicted or mutated miR-17 and miR-19 binding sites within Bmpr2, Smad5, and Smad4 3′-UTR sequences (see Supplementary Table S1) were cloned into pmirGLO according to manufacturer's instructions. SAOS-2 cells were maintained in Dulbecco's Modified Eagle's Media (DMEM) supplemented with 10% FBS, 4 mmol/L glutamine, and 100 units each of penicillin and streptomycin (GIBCO) at 37°C and 8% CO2. Cells were obtained from Dr. Emma Lees (DNAX) in September 1994 and tested by immunoblotting with antibodies to p53 (SC#100 and Sc#99) and Rb (Pharmingen, G3-245) before being frozen down in September 1994. Cells were seeded in 24-well plates (2.0 × 104 cells per well) and cotransfected the next day with 10 ng of pmirGLO reporter plasmids and 0 to 200 ng of MSCV-miR-17 ∼ 92-IRES-GFP, using Fugene HD (Promega). pCMV6 was used as control. Cells were lysed 48 hours later, and Dual-Luciferase Reporter Assays (Promega) were conducted on a Synergy 2 Biotek microplate reader.

Immunoblotting

P6 cerebella from wild-type and DKO mice were lysed using radioimmunoprecipitation assay (RIPA) buffer and proteins were extracted and 50 μg of proteins immunoblotted, as previously described (30), using antibodies to BMPR-II (BD Biosciences, 1:100) and actin (Santa Cruz Biotechnology, Inc; 1:2,000). The relative densities of Bmpr2 and actin bands were quantified using the gel analyzer plug-in on ImageJ64 (version 10.2; NIH) and normalized to relative Bmpr2 density in a NIH3T3 cell lysate (relative density = 1).

FISH

FISH for miR-17∼92 was conducted on 10 mouse SHH medulloblastomas [Ptch1+/−;Cdkn2c−/− (n = 6); Ptch1+/−;Trp53−/− (n = 4)] using the BAC clones RP23-132K20 for miR-17∼92 and RP23-35N6 for the chromosome 14 band A3 (30).

Immunofluorescence, data collection, and statistics

Medulloblastoma cells cultured on Matrigel-coated Lab-Tek-II CC2 4-Chamber Slides (Electron Microscopy Sciences) were treated with FAM-labeled tiny LNAs for 0.5, 1, 24, and 48 hours and fixed in 4% paraformaldehyde for 15 minutes at room temperature. Cells were counterstained with wheat germ agglutinin (WGA) conjugated to Alexa Fluor 594 and with 4′,6-diamidino-2-phenylindole, dihydrochloride (DAPI) according to Invitrogen's instructions. WGA is a lectin that binds to N-acetylglucosamine and N-acetylneuraminic acid (sialic acid) residues within the plasma membrane (31). Cells and tumor sections were stained with antibodies to Ki-67 (Leica, 1:1,000), FAM-AF594 (Invitrogen, 1:500), or GFP (Abcam, 1:500). Counterstained and immunostained cells were imaged using confocal microscopy (for details, see Supplementary Material). The number of FAM-, DAPI-, or Ki-67–positive cells was counted in each optical section using the cell counter plug-in on ImageJ64 (version 10.2; NIH). Four hundred or more cells, based on DAPI expression, were counted per treatment condition.

Macroscopic images of Evans blue dye within whole-mount mouse brain tumor and brain tumor sections were captured using the iSight camera on an iPhone 4S. Statistical significance was determined using the Student t test on GraphPad Prism software (version 5.0).

miR-19a is the highest expressed miRNA in SHH medulloblastomas

qRT-PCR analysis of miR-17, miR-18a, miR-19a, miR-20a, miR-19b-1, and miR-92-1 revealed that each member of the cluster was significantly overexpressed in purified medulloblastoma cells of independently derived medulloblastomas spontaneously arisen from 6 [Ptch1+/−;Cdkn2c−/−] mice (19) and 4 [Ptch1+/−;Trp53−/−] mice (18) compared with 1-month-old cerebellum from C57BL/6 mice. None of the 10 tumors showed amplification of miR-17∼92 (Supplementary Fig. S1A and S1B). miR-19a was the highest expressed in both SHH medulloblastoma mouse models followed by miR-20a in medulloblastomas from [Ptch1+/−;Cdkn2c−/−] and miR-17 in medulloblastomas from [Ptch1+/−;Trp53−/−] mice (Fig. 1B).

Efficient miRNA silencing within medulloblastoma cells by 8-mer LNA-anti-miRs

To assess tiny LNA uptake and subcellular localization, 8 μmol/L of FAM-labeled tiny LNAs were added directly to the culture medium. Medulloblastoma cells purified from 3 independently derived spontaneously occurring [Ptch1+/−;Cdkn2c−/−] tumors were incubated for different times before fixation and counterstained with WGA and DAPI. Confocal imaging revealed tiny LNAs in the nucleus and the cytosol of the cell soma and proximal neurites within 30 minutes after addition of tiny LNAs (Fig. 2A and B). FAM was observed in most cells treated with anti-miR-17, anti-mR-19, and scrambled LNA (Fig. 2C).

Figure 2.

8-mer LNA-anti-miRs are taken up by medulloblastoma cells. A, representative optical sections of FAM-labeled scrambled LNA, nuclear DAPI, and WGA staining of the plasma membrane using a ×10 objective. Scale bar, 20 μm. Localization of FAM-labeled tiny LNAs within the cell and not at the cell surface (see merged images). B, optical sections of FAM-labeled tiny LNAs and DAPI-counterstained cells in medulloblastoma cells treated with anti-miR-17 and anti-miR-19 using a ×63 objective. Scale bar, 5 μm. C, FAM-labeled and DAPI-counterstained medulloblastoma cells were counted to determine the percentage of fluorescently labeled cells. Error bars, SD.

Figure 2.

8-mer LNA-anti-miRs are taken up by medulloblastoma cells. A, representative optical sections of FAM-labeled scrambled LNA, nuclear DAPI, and WGA staining of the plasma membrane using a ×10 objective. Scale bar, 20 μm. Localization of FAM-labeled tiny LNAs within the cell and not at the cell surface (see merged images). B, optical sections of FAM-labeled tiny LNAs and DAPI-counterstained cells in medulloblastoma cells treated with anti-miR-17 and anti-miR-19 using a ×63 objective. Scale bar, 5 μm. C, FAM-labeled and DAPI-counterstained medulloblastoma cells were counted to determine the percentage of fluorescently labeled cells. Error bars, SD.

Close modal

To assess the specificity of the LNA-anti-miRs in inhibiting miRNA function, RNA was extracted from medulloblastoma cells treated with anti-miR-17 and anti-miR-19a for 48 hours followed by qRT-PCR analysis of miR-17 and miR-19a levels within each sample from 3 tumors. The relative miR-17 and miR-19a levels in anti-miR-17- and anti-miR-19–treated cells, respectively, were decreased in each tumor compared with cells untreated or treated with scrambled LNA (Supplementary Fig. S2A and S2B).

8-mer LNA-anti-miRs suppress medulloblastoma proliferation in vitro

To determine whether 8-mer anti-miRs led to suppression of medulloblastoma proliferation in vitro, medulloblastoma cells were treated for 48 hours with 8 μmol/L tiny LNAs based on a dose response (Supplementary Fig. S3A). Cells were harvested and counted. There was no significant change in the number of cells treated with the scrambled LNA compared with those left untreated. In contrast, we found significantly fewer cells after treatment with anti-miR-17, anti-miR-19, or anti-miR-17 with anti-miR-19 compared with untreated or scrambled LNA–treated controls; however, we did not observe an enhanced phenotype following treatment with both anti-miRs compared with either anti-miR alone (Supplementary Fig. S3B). Thus, each anti-miR was tested separately for the remainder of the experiments.

To assess whether the decrease in cell number was due to cell-cycle arrest or cell death, we measured proliferation by BrdUrd labeling and Ki-67 staining and apoptosis by Annexin V staining. Treatment of cells with anti-miR-17 or anti-miR-19 decreased the percentage of cells that incorporated BrdUrd and the number of Ki-67 immunoreactive cells without evidence of apoptosis (Supplementary Fig. S3C–S3E).

Pretreatment of medulloblastoma cells with 8-mer LNA-anti-miRs inhibits allograft proliferation

Tiny LNAs were added to 2 × 106 tumor cells from 4 individually derived spontaneous medulloblastomas during the 30-minute preplating step before injection into the flank of naïve recipient mice. Secondary tumors monitored by ultrasound imaging arose between 2 weeks and 1 month after transplant. Tumors from cells treated with anti-miR-17 or anti-miR-19 were significantly smaller in volume (Fig. 3A) and weight (Fig. 3B) than those untreated or scrambled LNA–treated cells.

Figure 3.

Pretreatment with 8-mer LNA-anti-miRs inhibits growth of flank allografts. A, line graph of tumor volume after transplant of cells left untreated (n = 5) or pretreated with scrambled LNA (n = 5), anti-miR-17 (n = 7), or anti-miR-19 (n = 5) during a 30-minute preplate. Imaging sessions began when tumors were palpable (day 0) and every week thereafter. B, bar graph of tumor weight following transplant of medulloblastoma cells left untreated or pretreated with tiny LNAs. C, relative levels of miR-17 and miR-19a seed families and miR-92-1 from RNA extracted from P6 cerebella of DKO mice and tumors derived from cells left untreated or pretreated with tiny LNAs. D, DKO; U, untreated; S, scrambled LNA; 17, anti-miR-17; 19, anti-miR-19. Error bars, SD. *, **, P ≤ 0.05 and P ≤ 0.01 compared with untreated; #, ##, ###, P ≤ 0.05, P ≤ 0.01, and P ≤ 0.001 compared with scrambled LNA.

Figure 3.

Pretreatment with 8-mer LNA-anti-miRs inhibits growth of flank allografts. A, line graph of tumor volume after transplant of cells left untreated (n = 5) or pretreated with scrambled LNA (n = 5), anti-miR-17 (n = 7), or anti-miR-19 (n = 5) during a 30-minute preplate. Imaging sessions began when tumors were palpable (day 0) and every week thereafter. B, bar graph of tumor weight following transplant of medulloblastoma cells left untreated or pretreated with tiny LNAs. C, relative levels of miR-17 and miR-19a seed families and miR-92-1 from RNA extracted from P6 cerebella of DKO mice and tumors derived from cells left untreated or pretreated with tiny LNAs. D, DKO; U, untreated; S, scrambled LNA; 17, anti-miR-17; 19, anti-miR-19. Error bars, SD. *, **, P ≤ 0.05 and P ≤ 0.01 compared with untreated; #, ##, ###, P ≤ 0.05, P ≤ 0.01, and P ≤ 0.001 compared with scrambled LNA.

Close modal

To determine whether the repression of flank tumor proliferation corresponded to a decrease in miRNA levels, RNAs from tumors were subjected to qRT-PCR. Tumors that arose from medulloblastoma cells treated with anti-miR-17 or anti-miR-19 exhibited a significant decrease in the relative levels of miR-17 and miR-19a seed sharing family members, respectively (Fig. 3C). The relative levels of miR-92, an miRNA that is not targeted by the 2 anti-miRs, were unchanged. Thus, 8-mer LNA-anti-miRs inhibit their cognate seed-sharing miRNA family members in vivo, as previously reported (23).

Intravenous treatment of mice with 8-mer LNA-anti-miRs inhibits tumor progression

As the levels of miRNAs encoded by miR-17∼92 were similar between tumors from [Ptch1+/−;Trp53−/−] and [Ptch1+/−;Cdkn2c−/−] mice, and the loss of Trp53 in [Ptch1+/−] mice facilitated tumor transplants compared with the [Ptch1+/−;Cdkn2c−/−] model, we used the [Ptch1+/−;Trp53−/−] medulloblastomas to test the effects of tiny LNAs in vivo. A total of 1 × 106 YFP and luciferase-expressing tumor cells originating from 3 spontaneously occurring medulloblastomas from [Ptch1+/−;Trp53−/−] mice (tumor A, B, and C) were transplanted into the left flank of immunocompromised recipient CD-1 mice. In all 3 cases, i.v. administration of anti-miR-17 or anti-miR-19 resulted in inhibition of flank tumor progression, as measured by luciferase activity (Fig. 4A–D), and decreased tumor weight (Fig. 4E). The inhibition of tumor progression was correlated with a significant decrease in the relative levels of miR-17 and miR-19a seed-sharing families (Fig. 4F).

Figure 4.

Systemically delivered 8-mer LNA-anti-miRs suppress tumor growth in flank allografts. Line graphs of luciferase activity within tumors from [Ptch1+/−;Trp53−/−] tumor A (A), tumor B (B), and tumor C (C) before and during i.v. administration of saline, scrambled LNA, anti-miR-17, or anti-miR-19. Green arrowheads, days of tiny LNA treatment following transplant (days 2, 6, 9, and 13). D, representative images of luciferase activity in resulting flank tumors from 3 [Ptch1+/−;Trp53−/−] medulloblastomas (A–C) before tiny LNA treatment (day 1) and after treatment (days 8 and 15). Sa, saline; Scr, scrambled LNA; 17, anti-miR-17; 19, anti-miR-19. Colored bars represent increasing radiance intensity from blue to red. E, bar graph of tumor weight following treatment with saline (n = 6), scrambled LNA (n = 7), anti-miR-17 (n = 9), or anti-miR-19 (n = 9). F, relative levels of miR-17 and miR-19a seed families and miR-92-1 from RNA from P6 cerebella of DKO mice and tumors originating from tumor-bearing mice treated with saline or tiny LNAs. D, DKO; C, saline; S, scrambled LNA; 17, anti-miR-17; 19, anti-miR-19. Error bars, SD. *, **, and ***, P ≤ 0.05, P ≤ 0.01, and P ≤ 0.001 compared with saline; #, ###, P ≤ 0.05 and P ≤ 0.001 compared with scrambled LNA. +, P ≤ 0.05 compared with anti-miR-19.

Figure 4.

Systemically delivered 8-mer LNA-anti-miRs suppress tumor growth in flank allografts. Line graphs of luciferase activity within tumors from [Ptch1+/−;Trp53−/−] tumor A (A), tumor B (B), and tumor C (C) before and during i.v. administration of saline, scrambled LNA, anti-miR-17, or anti-miR-19. Green arrowheads, days of tiny LNA treatment following transplant (days 2, 6, 9, and 13). D, representative images of luciferase activity in resulting flank tumors from 3 [Ptch1+/−;Trp53−/−] medulloblastomas (A–C) before tiny LNA treatment (day 1) and after treatment (days 8 and 15). Sa, saline; Scr, scrambled LNA; 17, anti-miR-17; 19, anti-miR-19. Colored bars represent increasing radiance intensity from blue to red. E, bar graph of tumor weight following treatment with saline (n = 6), scrambled LNA (n = 7), anti-miR-17 (n = 9), or anti-miR-19 (n = 9). F, relative levels of miR-17 and miR-19a seed families and miR-92-1 from RNA from P6 cerebella of DKO mice and tumors originating from tumor-bearing mice treated with saline or tiny LNAs. D, DKO; C, saline; S, scrambled LNA; 17, anti-miR-17; 19, anti-miR-19. Error bars, SD. *, **, and ***, P ≤ 0.05, P ≤ 0.01, and P ≤ 0.001 compared with saline; #, ###, P ≤ 0.05 and P ≤ 0.001 compared with scrambled LNA. +, P ≤ 0.05 compared with anti-miR-19.

Close modal

The blood–brain barrier is compromised following intracranial tumor cell transplantation

A major obstacle of using 8-mer LNA-anti-miRs for the treatment of brain tumors is their inability to cross the intact blood–brain barrier (BBB; ref. 23). Following intracranial transplant, Evans blue or FAM-labeled scrambled LNA were administered i.v. to 3 mice. We observed Evans blue dye accumulation around the implant site and tumor extravasation (Fig. 5A). FAM accumulation was similarly detected in tumors from three mice that received FAM-labeled scrambled LNA (Fig. 5B), suggesting that cranial implants compromise the BBB.

Figure 5.

Intravenously delivered LNA-anti-miRs suppress brain allograft growth. A, images of 3 mouse brains with Evans blue accumulation around the injection site (left) and within a representative tumor in a whole brain mount (middle, top) and a section (middle, bottom). Right panel is a ×5 zoom of the white boxed in area on the brain section image. Scale bars, 0.5 cm (left), 0.25 cm (middle), and 0.5 mm (right). B, images of YFP-expressing cells 2 weeks after transplant and FAM-labeled scrambled LNA 30 minutes (mouse 1 and 2) and 60 minutes (mouse 3) after injection. Scale bar, 25 μm, ×10 objective. C, line graph of luciferase activity within tumors before and during i.v. delivery of saline (n = 3), scrambled LNA (n = 4), anti-miR-17 (n = 5), or anti-miR-19 (n = 4). Green arrowheads, days of tiny LNA treatment after transplant (days 2, 6, and 9). D, images of luciferase activity within tumors before (day 1) and following saline and tiny LNAs administration (days 8, 12, and 15). Sa, saline; S, scrambled LNA; 17, anti-miR-17; and 19, anti-miR-19. Colored bars represent increasing radiance intensity from blue to red. E, Kaplan–Meijer plot of tumor-bearing mice treated with saline and tiny LNAs. F, relative levels of miR-17 and miR-19a seed families and miR-92-1 within RNA extracted from P6 cerebella of DKO mice and medulloblastomas originating from mice treated with saline or tiny LNAs. D, DKO; C, saline; S, scrambled LNA; 17, anti-miR-17; 19, anti-miR-19. Error bars, SD. *, ***, P ≤ 0.05 and P ≤ 0.001 versus saline. #, ##, ###, P ≤ 0.05, P ≤ 0.01, and P ≤ 0.001 versus scrambled LNA.

Figure 5.

Intravenously delivered LNA-anti-miRs suppress brain allograft growth. A, images of 3 mouse brains with Evans blue accumulation around the injection site (left) and within a representative tumor in a whole brain mount (middle, top) and a section (middle, bottom). Right panel is a ×5 zoom of the white boxed in area on the brain section image. Scale bars, 0.5 cm (left), 0.25 cm (middle), and 0.5 mm (right). B, images of YFP-expressing cells 2 weeks after transplant and FAM-labeled scrambled LNA 30 minutes (mouse 1 and 2) and 60 minutes (mouse 3) after injection. Scale bar, 25 μm, ×10 objective. C, line graph of luciferase activity within tumors before and during i.v. delivery of saline (n = 3), scrambled LNA (n = 4), anti-miR-17 (n = 5), or anti-miR-19 (n = 4). Green arrowheads, days of tiny LNA treatment after transplant (days 2, 6, and 9). D, images of luciferase activity within tumors before (day 1) and following saline and tiny LNAs administration (days 8, 12, and 15). Sa, saline; S, scrambled LNA; 17, anti-miR-17; and 19, anti-miR-19. Colored bars represent increasing radiance intensity from blue to red. E, Kaplan–Meijer plot of tumor-bearing mice treated with saline and tiny LNAs. F, relative levels of miR-17 and miR-19a seed families and miR-92-1 within RNA extracted from P6 cerebella of DKO mice and medulloblastomas originating from mice treated with saline or tiny LNAs. D, DKO; C, saline; S, scrambled LNA; 17, anti-miR-17; 19, anti-miR-19. Error bars, SD. *, ***, P ≤ 0.05 and P ≤ 0.001 versus saline. #, ##, ###, P ≤ 0.05, P ≤ 0.01, and P ≤ 0.001 versus scrambled LNA.

Close modal

8-mer LNA-anti-miRs inhibit medulloblastoma progression in the brain and increases survival

To determine whether treatment with 8-mer LNA-anti-miRs could be effective in suppressing the proliferation of medulloblastomas transplanted into the brain, we transplanted 0.5 × 106 YFP and luciferase-positive cells from one of the medulloblastomas from [Ptch1+/−;Trp53−/−] mice into the cortices of CD-1 mice. Tumor-bearing mice were treated i.v. with saline-formulated tiny LNAs, and tumor growth was assessed using bioluminescence imaging. Tumors that developed in the cortices of mice treated with anti-miR-17 or anti-miR-19 exhibited less luciferase activity (Fig. 5C and D) and led to a 6-day and 3- to 4-day increase in mean survival, respectively, compared with saline and scrambled LNA–treated controls (Fig. 5E). Mice treated with tiny LNAs did not show any apparent signs of toxicity-related illness such as weight loss. qRT-PCR analysis revealed that tumors that arose from mice treated with anti-miR-17 had significantly reduced levels of miR-17, whereas its seed-sharing miR levels were not as significantly affected. Tumors treated with anti-miR-19 showed no significant reduction of the relative levels of miR-19a seed-sharing miRs (Fig. 5F).

miR-17 and miR-19 target Bmpr2

The miR-17∼92 cluster targets SMAD2/3 (32) and SMAD4 (33), key effectors within the TGFβ signaling cascade, and BMPR2 within the BMP signaling cascade in human pulmonary arterial smooth muscle cells (34). Here, we tested whether miR-17 and miR-19 targeted members of the BMP signaling pathway, which dominantly and irreversibly induce the differentiation of SHH medulloblastomas (27). Using TargetScan, we found that Bmpr2, Smad5, and Smad4 contained predicted binding sites for miR-17 and miR-19a within their 3′-UTR.

We used the firefly–Renilla dual luciferase reporter assay to test whether miR-17∼92 induced posttranscriptional repression of Bmpr2, Smad5, or Smad4. Transfections were conducted in the SAOS-2 cell line due to its relatively low expression levels of the miR-17∼92 cluster (http://www.microrna.org). We observed a concentration-dependent repression of Bmpr2 luciferase reporter activity with increasing concentrations of miR-17∼92, which was not observed with Smad5 or Smad4 luciferase reporters (Supplementary Fig. S4A and S4B). Mutations in the miR-17 and miR-19a predicted binding sites of the Bmpr2 3′-UTR caused a derepression of luciferase activity (Fig. 6A), implicating Bmpr2 as a target of miR-17 and miR-19. These data were confirmed by Bmpr2 immunoblots of lysates from P6 cerebella from DKO and wild-type mice (Fig. 6B and C).

Figure 6.

miR-17 and miR-19 target Bmpr2 in medulloblastoma cells. A, bar graph of Bmpr2 3′-UTR reporter assay in SAOS-2 cells. B, immunoblot of Bmpr2 expression in lysates of P6 cerebella from wild type (lanes 1–3) and DKO mice (lanes 4–6). C, bar graph showing relative Bmpr2 density.

Figure 6.

miR-17 and miR-19 target Bmpr2 in medulloblastoma cells. A, bar graph of Bmpr2 3′-UTR reporter assay in SAOS-2 cells. B, immunoblot of Bmpr2 expression in lysates of P6 cerebella from wild type (lanes 1–3) and DKO mice (lanes 4–6). C, bar graph showing relative Bmpr2 density.

Close modal

We investigated the role of miR-17 and miR-19a seed-sharing family members in medulloblastoma proliferation by inhibiting their function using 8-mer LNA-modified anti-miRs directed against their seed sequences, designated as anti-miR-17 and anti-miR-19. We found miR-19a, miR-17, and miR-20a to be the highest expressed miRNAs from miR-17∼92 within medulloblastomas from [Ptch1+/−;Cdkn2c−/−] and [Ptch1+/−;Trp53−/−] mice. Interestingly, overexpression of miR-19a and miR-19b are both capable of inducing B-cell lymphoma (14, 15). The differential expression of individual miRNAs from this cluster is explained, in part, by the tertiary structure of the miR-17∼92 pri-miRNA. Chaulk and colleagues revealed that this pri-miRNA cluster adopts a compact globular tertiary structure containing the 3′-miRNA hairpins (miR-19b and miR-92) within a core enveloped by the 5′-miRNA hairpins (miR-17, miR-19a, miR-20a), thereby affecting the efficiency of Drosha processing and the regulation of target mRNAs (35).

Tiny LNAs passively and efficiently entered medulloblastoma cells without the need of transfection or electroporation. In vitro treatment of purified medulloblastoma cells with anti-miR-17 or anti-miR-19 decreased their proliferation compared with untreated or scrambled LNA–treated controls. The diminished proliferative capacity of tumor cells after anti-miR-17 treatment may reflect the induction of a subset of antiproliferative genes, such as those responsible for differentiation. We found that Bmpr2 was a target of miR-17, as previously published (34), and extended these data by including miR-19. However, CLIP/SEQ experiments will be necessary to further identify bona fide targets for the miRNAs encoded by the miR-17∼92 cluster family within SHH medulloblastoma cells (36).

Our transplantation experiments provide several insights when considering the development of miRNA-based therapies for treatment of SHH medulloblastomas. First, we previously found variability in the expression of miRNAs from miR-17∼92 within human and mouse SHH medulloblastomas (21), which correlated with the fact that not all SHH medulloblastomas from [Ptch1+/−;Cdkn2c−/−] mice gave rise to secondary tumors in flank transplants. Interestingly, the relative expression levels of miR-17 and miR-19a were at least 2 to 3 times higher in transplantable tumors compared with those that were not (Supplementary Fig. S5A). Tumor sensitivity to 8-mer LNA-anti-miRs was correlated with the expression levels of miR-17∼92 (Supplementary Fig. S5B). Second, tiny LNAs were present in transplantable tumors for at least 14 days following medulloblastoma cell pretreatment (Supplementary Fig. S6). These data are consistent with previous findings showing the in vivo stability of tiny LNAs up to 21 days (23). Finally, we observed that tumors derived from cells pretreated with anti-miR-17 were smaller than those originating from cells pretreated with anti-miR-19. Similar results were obtained when tiny LNAs were administered intravenously into immunocompromised mice carrying flank and cranial allografts. One possible explanation is that anti-miR-17 targets the seed region of 4 miRNAs (miR-17, 20a, 106b, 93), possibly exhibiting a broader effect on medulloblastoma miRNA-mediated regulatory networks than anti-miR-19, which only targets 2 miRNAs (miR-19a and 19b-1).

Although systemic delivery of anti-miR-21 shows high levels of uptake and accumulation in the kidney cortex, liver, lymph nodes, bone marrow, and spleen of mice (23, 37), one of the major obstacles of using antisense oligonucleotides, including tiny LNAs, in the treatment of brain tumors is their inefficient delivery across the BBB (23). In this intracranial transplant model, the BBB was clearly compromised. However, it is unclear whether the integrity of the BBB is disrupted in children with medulloblastoma, which would facilitate brain penetration (38, 39). Other strategies to achieve direct delivery to tumors might be considered, including continuous infusion of tiny LNAs into the ventricles using minipumps (40) or the use of nanoparticles.

As the use of LNA-anti-miRs in non-human primates and humans is efficacious and safe (41–45), our data suggest that 8-mer LNA-anti-miRs targeting miR-17∼92 and miR-106b∼25 may have therapeutic application for the treatment of human SHH medulloblastomas expressing high levels of miR-17∼92 (22). The therapeutic value of LNA-anti-miRs in medulloblastoma could be extended to other oncomiRs such as, miR-1204 encoded by PVT1 and frequently fused to MYC in G3 medulloblastomas (46) and miR-182, which promotes G3 medulloblastoma cell proliferation and migration (47) and leptomeningeal spread of non-SHH medulloblastomas (48).

S. Obad and S. Kauppinen are and were, respectively, employees of Santaris Pharma, a clinical stage biopharmaceutical company that develops RNA-based therapeutics.

Conception and design: B.L. Murphy, S. Obad, S. Kauppinen, M.F. Roussel

Development of methodology: B.L. Murphy, O. Ayrault, S. Kauppinen, M.F. Roussel

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): B.L. Murphy, L. Bihannic, F. Zindy, M.F. Roussel

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): B.L. Murphy, S. Obad, L. Bihannic, O. Ayrault, M.F. Roussel

Writing, review, and/or revision of the manuscript: B.L. Murphy, S. Obad, F. Zindy, S. Kauppinen, M.F. Roussel

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S. Kauppinen, M.F. Roussel

Study supervision: M.F. Roussel

The authors thank Charles J. Sherr, Daisuke Kawauchi, and all members of the laboratory for their help during these experiments; Mark Hatley for the generous gift of pCMV6 and technical help with 3′-UTR assays; Tyler Jacks and Andrea Ventura for generously providing conditional miR-17∼92 and miR-106b∼25-null mice; and Fedor Karginov, Andrea Ventura, and Gregory Hannon for criticisms of the manuscript. They also thank Sarah Robinson and Shelly Wilkerson for managing the mouse colony; Jose Grenet and Dana Farmer for plasmid purification; Drs. Richard Ashmun and Ann-Marie Hamilton-Easton for flow cytometric analysis; Monique Payton, Cameron Ogg and Dr. Christopher Calabrese for ultrasound image acquisition and measurement of tumor volume; John Gray for lentiviruses; Sarah Robinson, Monique Payton, Shantel Brown, and Brittney Perdue for tail vein injections; Melissa Johnson and Shantel Brown for cranial implants; Dr. Yannan Ouyang for confocal imaging assistance; and Marc and Virginia Valentine for FISH and analysis.

This work was funded in part by NIH grant CA-096832 (M.F. Roussel), a core grant CA02165-29 (M.F. Roussel), Children's Brain Tumor Foundation Award (M.F. Roussel), the George J. Mitchell Endowed Fellowship (B.L. Murphy), and the American Lebanese-Syrian Associated Charities (ALSAC) of St. Jude Children's Research Hospital.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Ellison
DW
. 
Childhood medulloblastoma: novel approaches to the classification of a heterogeneous disease
.
Acta Neuropathol
2010
;
120
:
305
16
.
2.
Taylor
MD
,
Northcott
PA
,
Korshunov
A
,
Remke
M
,
Cho
YJ
,
Clifford
SC
, et al
Molecular subgroups of medulloblastoma: the current consensus
.
Acta Neuropathol
2012
;
123
:
465
72
.
3.
Ris
MD
,
Packer
R
,
Goldwein
J
,
Jones-Wallace
D
,
Boyett
JM
. 
Intellectual outcome after reduced-dose radiation therapy plus adjuvant chemotherapy for medulloblastoma: a Children's Cancer Group study
.
J Clin Oncol
2001
;
19
:
3470
6
.
4.
Packer
RJ
,
Vezina
G
. 
Management of and prognosis with medulloblastoma: therapy at a crossroads
.
Arch Neurol
2008
;
65
:
1419
24
.
5.
Romer
J
,
Curran
T
. 
Targeting medulloblastoma: small-molecule inhibitors of the Sonic Hedgehog pathway as potential cancer therapeutics
.
Cancer Res
2005
;
65
:
4975
8
.
6.
Rudin
CM
,
Hann
CL
,
Laterra
J
,
Yauch
RL
,
Callahan
CA
,
Fu
L
, et al
Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449
.
N Engl J Med
2009
;
361
:
1173
8
.
7.
Kimura
H
,
Ng
JM
,
Curran
T
. 
Transient inhibition of the Hedgehog pathway in young mice causes permanent defects in bone structure
.
Cancer Cell
2008
;
13
:
249
60
.
8.
Metcalfe
C
,
de Sauvage
FJ
. 
Hedgehog fights back: mechanisms of acquired resistance against Smoothened antagonists
.
Cancer Res
2011
;
71
:
5057
61
.
9.
Calin
GA
,
Croce
CM
. 
MicroRNA signatures in human cancers. Nature reviews
.
Cancer
2006
;
6
:
857
66
.
10.
Lewis
BP
,
Shih
IH
,
Jones-Rhoades
MW
,
Bartel
DP
,
Burge
CB
. 
Prediction of mammalian microRNA targets
.
Cell
2003
;
115
:
787
98
.
11.
Ventura
A
,
Young
AG
,
Winslow
MM
,
Lintault
L
,
Meissner
A
,
Erkeland
SJ
, et al
Targeted deletion reveals essential and overlapping functions of the miR-17 through 92 family of miRNA clusters
.
Cell
2008
;
132
:
875
86
.
12.
Brennecke
J
,
Stark
A
,
Russell
RB
,
Cohen
SM
. 
Principles of microRNA-target recognition
.
PLoS Biol
2005
;
3
:
e85
.
13.
He
L
,
Thomson
JM
,
Hemann
MT
,
Hernando-Monge
E
,
Mu
D
,
Goodson
S
, et al
A microRNA polycistron as a potential human oncogene
.
Nature
2005
;
435
:
828
33
.
14.
Mu
P
,
Han
YC
,
Betel
D
,
Yao
E
,
Squatrito
M
,
Ogrodowski
P
, et al
Genetic dissection of the miR-17∼92 cluster of microRNAs in Myc-induced B-cell lymphomas
.
Genes Dev
2009
;
23
:
2806
11
.
15.
Olive
V
,
Bennett
MJ
,
Walker
JC
,
Ma
C
,
Jiang
I
,
Cordon-Cardo
C
, et al
miR-19 is a key oncogenic component of mir-17-92
.
Genes Dev
2009
;
23
:
2839
49
.
16.
Hallahan
AR
,
Pritchard
JI
,
Hansen
S
,
Benson
M
,
Stoeck
J
,
Hatton
BA
, et al
The SmoA1 mouse model reveals that notch signaling is critical for the growth and survival of sonic hedgehog-induced medulloblastomas
.
Cancer Res
2004
;
64
:
7794
800
.
17.
Taylor
MD
,
Liu
L
,
Raffel
C
,
Hui
CC
,
Mainprize
TG
,
Zhang
X
, et al
Mutations in SUFU predispose to medulloblastoma
.
Nat Genet
2002
;
31
:
306
10
.
18.
Wetmore
C
,
Eberhart
DE
,
Curran
T
. 
Loss of p53 but not ARF accelerates medulloblastoma in mice heterozygous for patched
.
Cancer Res
2001
;
61
:
513
6
.
19.
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
67
.
20.
Ayrault
O
,
Zindy
F
,
Rehg
J
,
Sherr
CJ
,
Roussel
MF
. 
Two tumor suppressors, p27Kip1 and patched-1, collaborate to prevent medulloblastoma
.
Mol Cancer Res.
2009
;
7
:
33
40
.
21.
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
7
.
22.
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
55
.
23.
Obad
S
,
dos Santos
CO
,
Petri
A
,
Heidenblad
M
,
Broom
O
,
Ruse
C
, et al
Silencing of microRNA families by seed-targeting tiny LNAs
.
Nat Genet
2011
;
43
:
371
8
.
24.
Stenvang
J
,
Petri
A
,
Lindow
M
,
Obad
S
,
Kauppinen
S
. 
Inhibition of microRNA function by antimiR oligonucleotides
.
Silence
2012
;
3
:
1
.
25.
Leucci
E
,
Zriwil
A
,
Gregersen
LH
,
Jensen
KT
,
Obad
S
,
Bellan
C
, et al
Inhibition of miR-9 de-represses HuR and DICER1 and impairs Hodgkin lymphoma tumour outgrowth
in vivo.
Oncogene
2012
;
31
:
5081
9
.
26.
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
.
27.
Zhao
H
,
Ayrault
O
,
Zindy
F
,
Kim
JH
,
Roussel
MF
. 
Post-transcriptional down-regulation of Atoh1/Math1 by bone morphogenic proteins suppresses medulloblastoma development
.
Genes Dev
2008
;
22
:
722
7
.
28.
Kaya
M
,
Palanduz
A
,
Kalayci
R
,
Kemikler
G
,
Simsek
G
,
Bilgic
B
, et al
Effects of lipopolysaccharide on the radiation-induced changes in the blood-brain barrier and the astrocytes
.
Brain Res
2004
;
1019
:
105
12
.
29.
Alves da Silva
JA
,
Oliveira
KC
,
Camillo
MA
. 
Gyroxin increases blood-brain barrier permeability to Evans blue dye in mice
.
Toxicon
2011
;
57
:
162
7
.
30.
Zindy
F
,
Uziel
T
,
Ayrault
O
,
Calabrese
C
,
Valentine
M
,
Rehg
JE
, et al
Genetic alterations in mouse medulloblastomas and generation of tumors de novo from primary cerebellar granule neuron precursors
.
Cancer Res
2007
;
67
:
2676
84
.
31.
Wright
CS
. 
Structural comparison of the two distinct sugar binding sites in wheat germ agglutinin isolectin II
.
J Mol Biol
1984
;
178
:
91
104
.
32.
Mestdagh
P
,
Bostrom
AK
,
Impens
F
,
Fredlund
E
,
Van Peer
G
,
De Antonellis
P
, et al
The miR-17-92 microRNA cluster regulates multiple components of the TGF-beta pathway in neuroblastoma
.
Mol Cell
2010
;
40
:
762
73
.
33.
Dews
M
,
Fox
JL
,
Hultine
S
,
Sundaram
P
,
Wang
W
,
Liu
YY
, et al
The myc-miR-17∼92 axis blunts TGF{beta} signaling and production of multiple TGF{beta}-dependent antiangiogenic factors
.
Cancer Res
2010
;
70
:
8233
46
.
34.
Brock
M
,
Samillan
VJ
,
Trenkmann
M
,
Schwarzwald
C
,
Ulrich
S
,
Gay
RE
, et al
AntagomiR directed against miR-20a restores functional BMPR2 signalling and prevents vascular remodelling in hypoxia-induced pulmonary hypertension
.
Eur Heart J.
2012 Mar 26
.
[Epub ahead of print]
.
35.
Chaulk
SG
,
Thede
GL
,
Kent
OA
,
Xu
Z
,
Gesner
EM
,
Veldhoen
RA
, et al
Role of pri-miRNA tertiary structure in miR-17∼92 miRNA biogenesis
.
RNA Biol
2011
;
8
:
1105
14
.
36.
Darnell
RB
. 
HITS-CLIP: panoramic views of protein-RNA regulation in living cells
.
Wiley Interdiscip Rev RNA
2010
;
1
:
266
86
.
37.
Garchow
BG
,
Bartulos Encinas
O
,
Leung
YT
,
Tsao
PY
,
Eisenberg
RA
,
Caricchio
R
, et al
Silencing of microRNA-21 in vivo ameliorates autoimmune splenomegaly in lupus mice
.
EMBO Mol Med
2011
;
3
:
605
15
.
38.
Barai
S
,
Bandopadhayaya
GP
,
Julka
PK
,
Kale
SS
,
Kumar
R
,
Malhotra
A
, et al
Evaluation of Tc99m-glucoheptonate for SPECT functional imaging of medulloblastoma
.
J Clin Neurosci
2005
;
12
:
36
8
.
39.
Warnke
PC
,
Kopitzki
K
,
Timmer
J
,
Ostertag
CB
. 
Capillary physiology of human medulloblastoma: impact on chemotherapy
.
Cancer
2006
;
107
:
2223
7
.
40.
Kocerha
J
,
Faghihi
MA
,
Lopez-Toledano
MA
,
Huang
J
,
Ramsey
AJ
,
Caron
MG
, et al
MicroRNA-219 modulates NMDA receptor-mediated neurobehavioral dysfunction
.
Proc Natl Acad Sci U S A
2009
;
106
:
3507
12
.
41.
Elmén
J
,
Lindow
M
,
Schütz
S
,
Lawrence
M
,
Petri
A
,
Obad
S
, et al
LNA-mediated microRNA silencing in non-human primates
.
Nature
2008
;
452
:
896
9
.
42.
Lanford
RE
,
Hildebrandt-Eriksen
ES
,
Petri
A
,
Persson
R
,
Lindow
M
,
Munk
ME
, et al
Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection
.
Science
2010
;
327
:
198
201
.
43.
Hildebrandt-Eriksen
ES
,
Aarup
V
,
Persson
R
,
Hansen
HF
,
Munk
ME
,
Ørum
H
. 
A locked nucleic acid oligonucleotide targeting microRNA 122 is well-tolerated in cynomolgus monkeys
.
Nucleic Acid Ther
2012
;
22
:
152
61
.
44.
Lindow
M
,
Kauppinen
S
. 
Discovering the first microRNA-targeted drug
.
J Cell Biol
2012
;
199
:
407
12
.
45.
Janssen
HLA
,
Reesink
HW
,
Lawitz
EJ
,
Zeuzem
S
,
Rodriguez-Torres
M
,
Patel
K
, et al
Treatment of HCV infection by targeting microRNA
.
N Engl J Med
2013
;
368
:
1685
94
.
46.
Northcott
PA
,
Shih
DJH
,
Peacock
J
,
Garzia
L
,
Morrissy
AS
,
Zichner
T
, et al
Subgroup-specific structural variation across 1,000 medulloblastoma genomes
.
Nature
2012
;
488
:
49
56
.
47.
Weeraratne
SD
,
Amani
V
,
Teider
N
,
Pierre-Francois
J
,
Winter
D
,
Kye
MJ
, et al
Pleiotropic effects of miR-183∼96∼182 converge to regulate cell survival, proliferation and migration in medulloblastoma
.
Acta Neuropathol
2012
;
123
:
539
52
.
48.
Bai
AHC
,
Milde
T
,
Remke
M
,
Rolli
CG
,
Hielscher
T
,
Cho
Y-J
, et al
MicroRNA-182 promotes leptomeningeal spread of non-sonic hedgehog-medulloblastoma
.
Acta Neuropathol
2012
;
123
:
529
38
.