Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Mol Oncol ; 17(9): 1784-1802, 2023 09.
Article in English | MEDLINE | ID: mdl-37341142

ABSTRACT

Children with Group 3 medulloblastoma (G3 MB) have a very poor prognosis, and many do not survive beyond 5 years after diagnosis. A factor that may contribute to this is the lack of available targeted therapy. Expression of protein lin-28 homolog B (LIN28B), a regulator of developmental timing, is upregulated in several cancers, including G3 MB, and is associated with worse survival in this disease. Here, we investigate the role of the LIN28B pathway in G3 MB and demonstrate that the LIN28B-lethal-7 (let-7; a microRNA that is a tumor suppressor)-lymphokine-activated killer T-cell-originated protein kinase (PBK; also known as PDZ-binding kinase) axis promotes G3 MB proliferation. LIN28B knockdown in G3-MB-patient-derived cell lines leads to a significant reduction in cell viability and proliferation in vitro and in prolonged survival of mice with orthotopic tumors. The LIN28 inhibitor N-methyl-N-[3-(3-methyl-1,2,4-triazolo[4,3-b]pyridazin-6-yl)phenyl]acetamide (1632) significantly reduces G3 MB cell growth and demonstrates efficacy in reducing tumor growth in mouse xenograft models. Inhibiting PBK using HI-TOPK-032 also results in a significant reduction in G3 MB cell viability and proliferation. Together, these results highlight a critical role for the LIN28B-let-7-PBK pathway in G3 MB and provide preliminary preclinical results for drugs targeting this pathway.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , MicroRNAs , Humans , Mice , Animals , Medulloblastoma/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Cell Proliferation/genetics , MicroRNAs/genetics , Cerebellar Neoplasms/genetics , Cell Line, Tumor , RNA-Binding Proteins/genetics
2.
Acta Neuropathol Commun ; 11(1): 62, 2023 04 08.
Article in English | MEDLINE | ID: mdl-37029430

ABSTRACT

Medulloblastoma (MB) is the most common pediatric brain malignancy and is divided into four molecularly distinct subgroups: WNT, Sonic Hedgehog (SHHp53mut and SHHp53wt), Group 3, and Group 4. Previous reports suggest that SHH MB features a unique tumor microenvironment compared with other MB groups. To better understand how SHH MB tumor cells interact with and potentially modify their microenvironment, we performed cytokine array analysis of culture media from freshly isolated MB patient tumor cells, spontaneous SHH MB mouse tumor cells and mouse and human MB cell lines. We found that the SHH MB cells produced elevated levels of IGFBP2 compared to non-SHH MBs. We confirmed these results using ELISA, western blotting, and immunofluorescence staining. IGFBP2 is a pleiotropic member of the IGFBP super-family with secreted and intracellular functions that can modulate tumor cell proliferation, metastasis, and drug resistance, but has been understudied in medulloblastoma. We found that IGFBP2 is required for SHH MB cell proliferation, colony formation, and cell migration, through promoting STAT3 activation and upregulation of epithelial to mesenchymal transition markers; indeed, ectopic STAT3 expression fully compensated for IGFBP2 knockdown in wound healing assays. Taken together, our findings reveal novel roles for IGFBP2 in SHH medulloblastoma growth and metastasis, which is associated with very poor prognosis, and they indicate an IGFBP2-STAT3 axis that could represent a novel therapeutic target in medulloblastoma.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Humans , Child , Animals , Mice , Medulloblastoma/metabolism , Hedgehog Proteins/metabolism , Epithelial-Mesenchymal Transition , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cerebellar Neoplasms/metabolism , Tumor Microenvironment , STAT3 Transcription Factor/metabolism
3.
Front Oncol ; 12: 903830, 2022.
Article in English | MEDLINE | ID: mdl-35747808

ABSTRACT

Medulloblastoma (MB) is the most common malignant brain tumor in children with standard of care consisting of surgery, radiation, and chemotherapy. Recent molecular profiling led to the identification of four molecularly distinct MB subgroups - Wingless (WNT), Sonic Hedgehog (SHH), Group 3, and Group 4. Despite genomic MB characterization and subsequent tumor stratification, clinical treatment paradigms are still largely driven by histology, degree of surgical resection, and presence or absence of metastasis rather than molecular profile. Patients usually undergo resection of their tumor followed by craniospinal radiation (CSI) and a 6 month to one-year multi-agent chemotherapeutic regimen. While there is clearly a need for development of targeted agents specific to the molecular alterations of each patient, targeting proteins responsible for DNA damage repair could have a broader impact regardless of molecular subgrouping. DNA damage response (DDR) protein inhibitors have recently emerged as targeted agents with potent activity as monotherapy or in combination in different cancers. Here we discuss the molecular underpinnings of genomic instability in MB and potential avenues for exploitation through DNA damage response inhibition.

4.
Sci Rep ; 9(1): 13611, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31541170

ABSTRACT

Medulloblastoma is a malignant pediatric tumor that arises from neural progenitors in the cerebellum. Despite a five-year survival rate of ~70%, nearly all patients incur adverse side effects from current treatment strategies that drastically impact quality of life. Roughly one-third of medulloblastoma are driven by aberrant activation of the Sonic Hedgehog (SHH) signaling pathway. However, the scarcity of genetic mutations in medulloblastoma has led to investigation of other mechanisms contributing to cancer pathogenicity including epigenetic regulation of gene expression. Here, we show that Helicase, Lymphoid Specific (HELLS), a chromatin remodeler with epigenetic functions including DNA methylation and histone modification, is induced by Sonic Hedgehog (SHH) in SHH-dependent cerebellar progenitor cells and the developing murine cerebella. HELLS is also up-regulated in mouse and human SHH medulloblastoma. Others have shown that HELLS activity generally results in a repressive chromatin state. Our results demonstrate that increased expression of HELLS in our experimental systems is regulated by the oncogenic transcriptional regulator YAP1 downstream of Smoothened, the positive transducer of SHH signaling. Elucidation of HELLS as one of the downstream effectors of the SHH pathway may lead to novel targets for precision therapeutics with the promise of better outcomes for SHH medulloblastoma patients.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , DNA Helicases/genetics , Medulloblastoma/genetics , Adaptor Proteins, Signal Transducing/genetics , Adult , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Cerebellar Neoplasms/pathology , Cerebellum/metabolism , Child , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , DNA Helicases/metabolism , Epigenesis, Genetic/genetics , Female , Hedgehog Proteins/metabolism , Hedgehog Proteins/physiology , Humans , Male , Medulloblastoma/metabolism , Mice , Neural Stem Cells/metabolism , Neurons/metabolism , Quality of Life , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Up-Regulation , YAP-Signaling Proteins
5.
Dev Cell ; 48(2): 129-130, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30695693

ABSTRACT

In the developing cerebellum, Sonic hedgehog (SHH) signaling is required for expansion of cerebellar granule neural progenitors, proposed to be cells-of-origin for the SHH-driven pediatric brain tumor medulloblastoma. In this issue of Developmental Cell, Chang et al. (2019) show that the transcription factor Atoh1/MATH1 regulates primary cilium formation, enabling SHH signaling.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Basic Helix-Loop-Helix Transcription Factors , Cell Proliferation , Cerebellum , Child , Cilia , Hedgehog Proteins , Humans , Neurons
6.
Mol Cancer Res ; 17(1): 186-198, 2019 01.
Article in English | MEDLINE | ID: mdl-30224541

ABSTRACT

Medulloblastomas, the most common malignant pediatric brain tumors, have been genetically defined into four subclasses, namely WNT-activated, Sonic Hedgehog (SHH)-activated, Group 3, and Group 4. Approximately 30% of medulloblastomas have aberrant SHH signaling and thus are referred to as SHH-activated medulloblastoma. The tumor suppressor gene TP53 has been recently recognized as a prognostic marker for patients with SHH-activated medulloblastoma; patients with mutant TP53 have a significantly worse outcome than those with wild-type TP53. It remains unknown whether p53 activity is impaired in SHH-activated, wild-type TP53 medulloblastoma, which is about 80% of the SHH-activated medulloblastomas. Utilizing the homozygous NeuroD2:SmoA1 mouse model with wild-type Trp53, which recapitulates human SHH-activated medulloblastoma, it was discovered that the endogenous Inhibitor 2 of Protein Phosphatase 2A (SET/I2PP2A) suppresses p53 function by promoting accumulation of phospho-MDM2 (S166), an active form of MDM2 that negatively regulates p53. Knockdown of I2PP2A in SmoA1 primary medulloblastoma cells reduced viability and proliferation in a p53-dependent manner, indicating the oncogenic role of I2PP2A. Importantly, this mechanism is conserved in the human medulloblastoma cell line ONS76 with wild-type TP53. Taken together, these findings indicate that p53 activity is inhibited by I2PP2A upstream of PP2A in SHH-activated and TP53-wildtype medulloblastomas. IMPLICATIONS: This study suggests that I2PP2A represents a novel therapeutic option and its targeting could improve the effectiveness of current therapeutic regimens for SHH-activated or other subclasses of medulloblastoma with wild-type TP53.


Subject(s)
Cerebellar Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Hedgehog Proteins/metabolism , Histone Chaperones/metabolism , Medulloblastoma/metabolism , Tumor Suppressor Protein p53/metabolism , Adult , Animals , Cell Line, Tumor , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Histone Chaperones/antagonists & inhibitors , Histone Chaperones/genetics , Humans , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Peptides/pharmacology , Tumor Suppressor Protein p53/genetics , Up-Regulation
7.
Mol Cancer Res ; 14(1): 114-24, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26446920

ABSTRACT

UNLABELLED: Sonic hedgehog (Shh) signaling is closely coupled with bioenergetics of medulloblastoma, the most common malignant pediatric brain tumor. Shh-associated medulloblastoma arises from cerebellar granule neuron precursors (CGNP), a neural progenitor whose developmental expansion requires signaling by Shh, a ligand secreted by the neighboring Purkinje neurons. Previous observations show that Shh signaling inhibits fatty acid oxidation although driving increased fatty acid synthesis. Proliferating CGNPs and mouse Shh medulloblastomas feature high levels of glycolytic enzymes in vivo and in vitro. Because both of these metabolic processes are closely linked to mitochondrial bioenergetics, the role of Shh signaling in mitochondrial biogenesis was investigated. This report uncovers a surprising decrease in mitochondrial membrane potential (MMP) and overall ATP production in CGNPs exposed to Shh, consistent with increased glycolysis resulting in high intracellular acidity, leading to mitochondrial fragmentation. Ultrastructural examination of mitochondria revealed a spherical shape in Shh-treated cells, in contrast to the elongated appearance in vehicle-treated postmitotic cells. Expression of mitofusin 1 and 2 was reduced in these cells, although their ectopic expression restored the MMP to the nonproliferating state and the morphology to a fused, interconnected state. Mouse Shh medulloblastoma cells featured drastically impaired mitochondrial morphology, restoration of which by ectopic mitofusin expression was also associated with a decrease in the expression of Cyclin D2 protein, a marker for proliferation. IMPLICATIONS: This report exposes a novel role for Shh in regulating mitochondrial dynamics and rescue of the metabolic profile of tumor cells to that of nontransformed, nonproliferating cells and represents a potential avenue for development of medulloblastoma therapeutics.


Subject(s)
GTP Phosphohydrolases/metabolism , Hedgehog Proteins/metabolism , Mitophagy , Neural Stem Cells/metabolism , Animals , Animals, Newborn , Cell Proliferation , Cells, Cultured , Cerebellar Neoplasms , GTP Phosphohydrolases/genetics , Glycolysis , Medulloblastoma , Membrane Potential, Mitochondrial , Mice , Mitochondria/ultrastructure , Neoplasms, Experimental , Neural Stem Cells/cytology
8.
Dev Neurobiol ; 72(6): 789-804, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22095825

ABSTRACT

Precise regulation of neuroprogenitor cell proliferation and differentiation is required for successful brain development, but the factors that contribute to this are only incompletely understood. The transcription factor ATF5 promotes proliferation of cerebral cortical neuroprogenitor cells and its down regulation permits their differentiation. Here, we examine the expression and regulation of ATF5 in cerebellar granule neuron progenitor cells (CGNPs) as well as the role of ATF5 in the transition of CGNPs to postmitotic cerebellar granule neurons (GCNs). We find that ATF5 is expressed by proliferating CGNPs in both the embryonic and postnatal cerebellar external granule layer (EGL) and in the rhombic lip, the embryonic structure from which the EGL arises. In contrast, ATF5 is undetectable in postmitotic GCNs. In highly enriched dissociated cultures of CGNPs and CGNs, ATF5 is expressed only in CGNPs. Constitutive ATF5 expression in CGNPs does not affect their proliferation or exit from the cell cycle. In contrast, in presence of sonic hedgehog (Shh), a mitogen for CGNPs, constitutively expressed ATF5 promotes CGNP proliferation and delays their cell cycle exit and differentiation. Conversely, ATF5 loss-of-function conferred by a dominant-negative form of ATF5 significantly diminishes Shh-stimulated CGNP proliferation and promotes differentiation. In parallel with its stimulation of CGNP proliferation, Shh enhances ATF5 expression by what appeared to be a posttranscriptional mechanism involving protein stabilization. These findings indicate a reciprocal interaction between ATF5 and Shh in which Shh stimulates ATF5 expression and in which ATF5 contributes to Shh-stimulated CGNP expansion.


Subject(s)
Activating Transcription Factors/metabolism , Cell Proliferation , Cerebellum/cytology , Hedgehog Proteins/metabolism , Neural Stem Cells/cytology , Neurons/cytology , Activating Transcription Factors/genetics , Animals , Cells, Cultured , Cerebellum/metabolism , Gene Expression Regulation , Hedgehog Proteins/genetics , Mice , Neural Stem Cells/metabolism , Neurons/metabolism , Signal Transduction
9.
Cell Cycle ; 9(21): 4307-14, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-21051932

ABSTRACT

Medulloblastoma, a brain tumor arising in the cerebellum, is the most common solid childhood malignancy. the current standard of care for medulloblastoma leaves survivors with life-long side effects. Gaining insight into mechanisms regulating transformation of medulloblastoma cells-of-origin may lead to development of better treatments for these tumors. Cerebellar granule neuron precursors (CGNps) are proposed cells-of-origin for certain classes of medulloblastoma, specifically those marked by aberrant Sonic hedgehog (Shh) signaling pathway activation. CGNps require signaling by Shh for proliferation during brain development. In mitogen-stimulated cells, nuclear localized cyclin dependent kinase (cdk) inhibitor p27 (Kip1) functions as a checkpoint control at the G1- to S-phase transition by inhibiting cdk2. Recent studies have suggested cytoplasmically localized p27(Kip1) acquires oncogenic functions. Here, we show that p27(Kip1) is cytoplasmically localized in CGNps and mouse Shh-mediated medulloblastomas. transgenic mice bearing an activating mutation in the Shh pathway and lacking one or both p27(Kip1) alleles have accelerated tumor incidence compared to mice bearing both p27(Kip1) alleles. Interestingly, mice heterozygous for p27(Kip1) have decreased survival latency compared to p27(Kip1)-null animals. our data indicate that this may reflect the requirement for at least one copy of p27(Kip1) for recruiting cyclin D/cdk4/6 to promote cell cycle progression yet insufficient expression in the heterozygous or null state to inhibit cyclin E/cdk2. Finally, we find that mis-localized p27(Kip1) may play a positive role in motility in medulloblastoma cells. Together, our data indicate that the dosage of p27(Kip1) plays a role in cell cycle progression and tumor suppression in Shh-mediated medulloblastoma expansion.


Subject(s)
Cerebellar Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Hedgehog Proteins/metabolism , Medulloblastoma/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cells, Cultured , Cyclin D/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p27/analysis , Cyclin-Dependent Kinase Inhibitor p27/genetics , G1 Phase , Male , Mice , Mice, Transgenic , S Phase
10.
Cell Cycle ; 9(19): 4013-24, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20935513

ABSTRACT

Development of the cerebellum, a brain region regulating posture and coordination, occurs post-natally and is marked by rapid proliferation of granule neuron precursors (CGNPs), stimulated by mitogenic Sonic hedgehog (Shh) signaling. ß-Arrestin (ßArr) proteins play important roles downstream of Smoothened, the Shh signal transducer. However, whether Shh regulates ßArrs and what role they play in Shh-driven CGNP proliferation remains to be determined. Here, we report that Shh induces ßArr1 accumulation and localization to the nucleus, where it participates in enhancing expression of the cyclin dependent kinase (cdk) inhibitor p27, whose accumulation eventually drives CGNP cell cycle exit. ßArr1 knockdown enhances CGNP proliferation and reduces p27 expression. Thus, Shh-mediated ßArr1 induction represents a novel negative feedback loop within the Shh mitogenic pathway, such that ongoing Shh signaling, while required for CGNPs to proliferate, also sets up a cell-intrinsic clock programming their ultimate exit from the cell cycle.


Subject(s)
Arrestins/metabolism , Cell Cycle/physiology , Hedgehog Proteins/metabolism , Mitosis/physiology , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Signal Transduction/physiology , Animals , Arrestins/genetics , Cells, Cultured , Cerebellum/cytology , Cerebellum/growth & development , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Hedgehog Proteins/genetics , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Promoter Regions, Genetic , beta-Arrestin 1 , beta-Arrestins , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
11.
Cell Cycle ; 9(3): 456-9, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20081363

ABSTRACT

Certain types of medulloblastoma, the most common solid pediatric cancer, are proposed to arise from neural precursors known as cerebellar granule neuron precursors (CGNPs), which require signaling by Sonic hedgehog (Shh) and insulin-like growth factor (IGF) for their proliferation and survival. Aberrant activity of these pathways is implicated in medulloblastoma. IGF activates the mammalian Target of Rapamycin (mTOR), a growth-promoting kinase normally kept in check by the tumor suppressive Tuberous Sclerosis Complex (TSC), comprised of TSC1 and TSC2. TSC also counteracts proliferation by stabilizing the cyclin-dependent kinase inhibitor p27(Kip1), preventing progression through G(1)- to S-phase of the cell cycle. We reported that mice with impaired TSC activity show increased susceptibility to Shh-mediated medulloblastoma. CGNPs and tumors from these mice display increased proliferation, mTOR pathway activation, glycogen synthase kinase-3 (GSK-3) alpha/beta inactivation, and atypical p27(Kip1) cytoplasmic localization. GSK-3alpha/beta inactivation was mTOR-dependent, whereas p27(Kip1) localization was uncoupled from mTOR, and was instead regulated by TSC2. These results provide insight into the molecular 'hardwiring' of the mitogenic network downstream of Shh signaling and emphasize the separate yet synergistic effects regulated by the TSC complex in (1) fueling proliferation through mTOR activation/GSK-3alpha/beta inactivation and (2) compromising checkpoint mechanisms via TSC2-dependent p27(Kip1) nuclear exclusion. Future medulloblastoma therapies targeting Shh signaling can be developed to selectively modulate these activities, to restore checkpoint control and attenuate uncontrolled hyperproliferation.


Subject(s)
Gene Silencing , Hedgehog Proteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Humans , Medulloblastoma/pathology , Mice , Models, Biological , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein
13.
Genes Dev ; 23(23): 2729-41, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19952108

ABSTRACT

Medulloblastoma is the most common solid malignancy of childhood, with treatment side effects reducing survivors' quality of life and lethality being associated with tumor recurrence. Activation of the Sonic hedgehog (Shh) signaling pathway is implicated in human medulloblastomas. Cerebellar granule neuron precursors (CGNPs) depend on signaling by the morphogen Shh for expansion during development, and have been suggested as a cell of origin for certain medulloblastomas. Mechanisms contributing to Shh pathway-mediated proliferation and transformation remain poorly understood. We investigated interactions between Shh signaling and the recently described tumor-suppressive Hippo pathway in the developing brain and medulloblastomas. We report up-regulation of the oncogenic transcriptional coactivator yes-associated protein 1 (YAP1), which is negatively regulated by the Hippo pathway, in human medulloblastomas with aberrant Shh signaling. Consistent with conserved mechanisms between brain tumorigenesis and development, Shh induces YAP1 expression in CGNPs. Shh also promotes YAP1 nuclear localization in CGNPs, and YAP1 can drive CGNP proliferation. Furthermore, YAP1 is found in cells of the perivascular niche, where proposed tumor-repopulating cells reside. Post-irradiation, YAP1 was found in newly growing tumor cells. These findings implicate YAP1 as a new Shh effector that may be targeted by medulloblastoma therapies aimed at eliminating medulloblastoma recurrence.


Subject(s)
Cerebellar Neoplasms/metabolism , Hedgehog Proteins/metabolism , Medulloblastoma/metabolism , Neurons/cytology , Neurons/metabolism , Up-Regulation , Animals , Cell Proliferation , Cells, Cultured , Cerebellar Neoplasms/pathology , Humans , Insulin Receptor Substrate Proteins/metabolism , Medulloblastoma/pathology , Mice , Protein Transport , Signal Transduction , Transcription Factors/metabolism
14.
Cell Cycle ; 8(24): 4049-54, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19901543

ABSTRACT

MicroRNAS (miRNAs) are small endogenous non-coding RNAs that play important roles in many different biological processes including proliferation, differentiation and apoptosis through silencing of target genes. Emerging evidence indicates that miRNAs are key players in mammalian development that, when altered, contribute to tumorigenesis. However, only a few studies to date have focused on the role of miRNAs in medulloblastoma, the most common malignant pediatric brain tumor. These tumors arise in the cerebellum and may attribute their origins to deregulated proliferation of neural progenitor cells during development. Understanding the interplay between normal brain development and medulloblastoma pathogenesis is necessary in order for more efficient, less toxic targeted therapies to be developed and implemented. MiRNA expression profiling of both mouse and human medulloblastomas has led to the identification of signatures correlating with the molecular subgroups of medulloblastoma, tumor diagnosis and response to treatment, as well as novel targets of potential clinical relevance. This review summarizes the recent miRNA literature in both medulloblastoma and normal brain development.


Subject(s)
Brain/embryology , Brain/physiology , Cerebellar Neoplasms/genetics , Medulloblastoma/genetics , MicroRNAs/physiology , Animals , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Humans
15.
Cancer Res ; 69(18): 7224-34, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19738049

ABSTRACT

During development, proliferation of cerebellar granule neuron precursors (CGNP), candidate cells-of-origin for the pediatric brain tumor medulloblastoma, requires signaling by Sonic hedgehog (Shh) and insulin-like growth factor (IGF), the pathways of which are also implicated in medulloblastoma. One of the consequences of IGF signaling is inactivation of the mammalian target of rapamycin (mTOR)-suppressing tuberous sclerosis complex (TSC), comprised of TSC1 and TSC2, leading to increased mRNA translation. We show that mice, in which TSC function is impaired, display increased mTOR pathway activation, enhanced CGNP proliferation, glycogen synthase kinase-3 alpha/beta (GSK-3 alpha/beta) inactivation, and cytoplasmic localization of the cyclin-dependent kinase inhibitor p27(Kip1), which has been proposed to cause its inactivation or gain of oncogenic functions. We observed the same characteristics in wild-type primary cultures of CGNPs in which TSC1 and/or TSC2 were knocked down, and in mouse medulloblastomas induced by ectopic Shh pathway activation. Moreover, Shh-induced mouse medulloblastomas manifested Akt-mediated TSC2 inactivation, and the mutant TSC2 allele synergized with aberrant Shh signaling to increase medulloblastoma incidence in mice. Driving exogenous TSC2 expression in Shh-induced medulloblastoma cells corrected p27(Kip1) localization and reduced proliferation. GSK-3 alpha/beta inactivation in the tumors in vivo and in primary CGNP cultures was mTOR-dependent, whereas p27(Kip1) cytoplasmic localization was regulated upstream of mTOR by TSC2. These results indicate that a balance between Shh mitogenic signaling and TSC function regulating new protein synthesis and cyclin-dependent kinase inhibition is essential for the normal development and prevention of tumor formation or expansion.


Subject(s)
Carrier Proteins/metabolism , Cerebrum/growth & development , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Medulloblastoma/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Growth Processes/physiology , Cerebrum/metabolism , Hedgehog Proteins/metabolism , Medulloblastoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein
16.
Cancer Res ; 69(8): 3249-55, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19351822

ABSTRACT

Medulloblastoma is the most common malignant pediatric brain tumor, and mechanisms underlying its development are poorly understood. We identified recurrent amplification of the miR-17/92 polycistron proto-oncogene in 6% of pediatric medulloblastomas by high-resolution single-nucleotide polymorphism genotyping arrays and subsequent interphase fluorescence in situ hybridization on a human medulloblastoma tissue microarray. Profiling the expression of 427 mature microRNAs (miRNA) in a series of 90 primary human medulloblastomas revealed that components of the miR-17/92 polycistron are the most highly up-regulated miRNAs in medulloblastoma. Expression of miR-17/92 was highest in the subgroup of medulloblastomas associated with activation of the sonic hedgehog (Shh) signaling pathway compared with other subgroups of medulloblastoma. Medulloblastomas in which miR-17/92 was up-regulated also had elevated levels of MYC/MYCN expression. Consistent with its regulation by Shh, we observed that Shh treatment of primary cerebellar granule neuron precursors (CGNP), proposed cells of origin for the Shh-associated medulloblastomas, resulted in increased miR-17/92 expression. In CGNPs, the Shh effector N-myc, but not Gli1, induced miR-17/92 expression. Ectopic miR-17/92 expression in CGNPs synergized with exogenous Shh to increase proliferation and also enabled them to proliferate in the absence of Shh. We conclude that miR-17/92 is a positive effector of Shh-mediated proliferation and that aberrant expression/amplification of this miR confers a growth advantage to medulloblastomas.


Subject(s)
Cerebellar Neoplasms/genetics , Hedgehog Proteins/metabolism , Medulloblastoma/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins c-myc/genetics , Adult , Animals , Cell Growth Processes/genetics , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Gene Amplification , Hedgehog Proteins/genetics , Hedgehog Proteins/pharmacology , Humans , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice , MicroRNAs/biosynthesis , Multigene Family , Neurons/drug effects , Neurons/pathology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/biosynthesis , Signal Transduction , Stem Cells/drug effects , Stem Cells/pathology , Up-Regulation
17.
Cancer Cell ; 15(1): 67-78, 2009 Jan 06.
Article in English | MEDLINE | ID: mdl-19111882

ABSTRACT

In human neuroblastoma, amplification of the MYCN gene predicts poor prognosis and resistance to therapy. In a shRNA screen of genes that are highly expressed in MYCN-amplified tumors, we have identified AURKA as a gene that is required for the growth of MYCN-amplified neuroblastoma cells but largely dispensable for cells lacking amplified MYCN. Aurora A has a critical function in regulating turnover of the N-Myc protein. Degradation of N-Myc requires sequential phosphorylation by cyclin B/Cdk1 and Gsk3. N-Myc is therefore degraded during mitosis in response to low levels of PI3-kinase activity. Aurora A interacts with both N-Myc and the SCF(Fbxw7) ubiquitin ligase that ubiquitinates N-Myc and counteracts degradation of N-Myc, thereby uncoupling N-Myc stability from growth factor-dependent signals.


Subject(s)
Neuroblastoma/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Aurora Kinase A , Aurora Kinases , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Protein Binding , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , Ubiquitin-Protein Ligases/metabolism
18.
Development ; 135(19): 3291-300, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18755774

ABSTRACT

Sonic hedgehog (SHH) and insulin-like growth factor (IGF) signaling are essential for development of many tissues and are implicated in medulloblastoma, the most common solid pediatric malignancy. Cerebellar granule neuron precursors (CGNPs), proposed cells-of-origin for specific classes of medulloblastomas, require SHH and IGF signaling for proliferation and survival during development of the cerebellum. We asked whether SHH regulates IGF pathway components in proliferating CGNPs. We report that SHH-treated CGNPs showed increased levels of insulin receptor substrate 1 (IRS1) protein, which was also present in the germinal layer of the developing mouse cerebellum and in mouse SHH-induced medulloblastomas. Previous roles for IRS1, an oncogenic protein that is essential for IGF-mediated proliferation in other cell types, have not been described in SHH-mediated CGNP proliferation. We found that IRS1 overexpression can maintain CGNP proliferation in the absence of SHH. Furthermore, lentivirus-mediated knock down experiments have shown that IRS1 activity is required for CGNP proliferation in slice explants and dissociated cultures. Contrary to traditional models for SHH signaling that focus on gene transcription, SHH stimulation does not regulate Irs1 transcription but rather stabilizes IRS1 protein by interfering with mTOR-dependent IRS1 turnover and possibly affects Irs1 mRNA translation. Thus, we have identified IRS1 as a novel effector of SHH mitogenic signaling that may serve as a future target for medulloblastoma therapies. Our findings also indicate a previously unreported interaction between the SHH and mTOR pathways, and provide an example of a non-classical means for SHH-mediated protein regulation during development.


Subject(s)
Cerebellum/cytology , Cerebellum/metabolism , Hedgehog Proteins/metabolism , Insulin Receptor Substrate Proteins/metabolism , Animals , Base Sequence , Carrier Proteins/metabolism , Cell Proliferation , Cells, Cultured , Cerebellar Neoplasms/etiology , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Cerebellum/growth & development , DNA Primers/genetics , Hedgehog Proteins/genetics , Insulin Receptor Substrate Proteins/genetics , Medulloblastoma/etiology , Medulloblastoma/genetics , Medulloblastoma/metabolism , Mice , Mice, Mutant Strains , Mitosis , Models, Neurological , Neurons/cytology , Neurons/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , TOR Serine-Threonine Kinases , Up-Regulation
19.
Cancer Res ; 66(17): 8655-61, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951180

ABSTRACT

We examined the genetic requirements for the Myc family of oncogenes in normal Sonic hedgehog (Shh)-mediated cerebellar granule neuronal precursor (GNP) expansion and in Shh pathway-induced medulloblastoma formation. In GNP-enriched cultures derived from N-myc(Fl/Fl) and c-myc(Fl/Fl) mice, disruption of N-myc, but not c-myc, inhibited the proliferative response to Shh. Conditional deletion of c-myc revealed that, although it is necessary for the general regulation of brain growth, it is less important for cerebellar development and GNP expansion than N-myc. In vivo analysis of compound mutants carrying the conditional N-myc null and the activated Smoothened (ND2:SmoA1) alleles showed, that although granule cells expressing the ND2:SmoA1 transgene are present in the N-myc null cerebellum, no hyperproliferation or tumor formation was detected. Taken together, these findings provide in vivo evidence that N-myc acts downstream of Shh/Smo signaling during GNP proliferation and that N-myc is required for medulloblastoma genesis even in the presence of constitutively active signaling from the Shh pathway.


Subject(s)
Cerebellar Neoplasms/genetics , Cerebellum/cytology , Genes, myc , Hedgehog Proteins/physiology , Medulloblastoma/genetics , Animals , Cell Division , Cerebellar Neoplasms/pathology , Cerebellum/pathology , Child , Humans , Kinetics , Medulloblastoma/pathology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction
20.
Cell Cycle ; 5(1): 47-52, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16322694

ABSTRACT

Signaling by the sonic hedgehog (Shh) pathway is essential for neural precursor population expansion during normal central nervous system (CNS) development, and is implicated in the childhood brain tumor, medulloblastoma. The proto-oncogene N-myc plays essential roles as a downstream effector of Shh proliferative effects in neural precursors of the cerebellum, where medulloblastomas arise. It is likely that N-Myc has analogous functions in medulloblastomas and other CNS tumors where it is highly expressed due to altered regulation or gene amplification. Myc destabilization occurs in response to phosphorylation by GSK-3beta. N-Myc degradation is required for cerebellar neural precursors to exit the cell cycle. During mitosis in cerebellar neural precursors, levels of N-Myc primed for phosphorylation by GSK-3beta increase, due to cdk1 complex activity towards N-Myc. GSK-3beta is kept in check by insulin-like growth factor signaling, which also plays critical roles in brain development and cancer. These findings indicate that therapeutic strategies targeting N-myc and the IGF pathway might be effective against medulloblastoma.


Subject(s)
Cell Differentiation , Glycogen Synthase Kinase 3/metabolism , Neurons/cytology , Neurons/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Trans-Activators/metabolism , Animals , Hedgehog Proteins , Humans , Proto-Oncogene Mas , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...