Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nat Commun ; 10(1): 3731, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31427603

ABSTRACT

Pilocytic astrocytoma (PA), the most common childhood brain tumor, is a low-grade glioma with a single driver BRAF rearrangement. Here, we perform scRNAseq in six PAs using methods that enabled detection of the rearrangement. When compared to higher-grade gliomas, a strikingly higher proportion of the PA cancer cells exhibit a differentiated, astrocyte-like phenotype. A smaller proportion of cells exhibit a progenitor-like phenotype with evidence of proliferation. These express a mitogen-activated protein kinase (MAPK) programme that was absent from higher-grade gliomas. Immune cells, especially microglia, comprise 40% of all cells in the PAs and account for differences in bulk expression profiles between tumor locations and subtypes. These data indicate that MAPK signaling is restricted to relatively undifferentiated cancer cells in PA, with implications for investigational therapies directed at this pathway.


Subject(s)
Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/pathology , Neural Stem Cells/cytology , Proto-Oncogene Proteins B-raf/genetics , Animals , Brain Neoplasms/genetics , Humans , MAP Kinase Signaling System/genetics , Mice , Microglia/pathology , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Oligodendroglia/cytology , Oncogene Proteins, Fusion/metabolism , Tumor Cells, Cultured
2.
Cancer Res ; 78(14): 4007-4021, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29760046

ABSTRACT

Aberrant chromatin remodeling and activation of the PI3K pathway have been identified as important mediators of pediatric high-grade glioma (pHGG) and diffuse intrinsic pontine glioma (DIPG) pathogenesis. As inhibition of these pathways are promising therapeutic avenues and radiation is the only modality to prolong survival of patients with DIPG, we sought to explore radiosensitizing functions of such inhibition and to explore mechanisms of action of such agents. Here, we demonstrate that combined treatment with radiotherapy and CUDC-907, a novel first-in-class dual inhibitor of histone deacetylases (HDAC) and PI3K, evokes a potent cytotoxic response in pHGG and DIPG models. CUDC-907 modulated DNA damage response by inhibiting radiation-induced DNA repair pathways including homologous recombination and nonhomologous end joining. The radiosensitizing effects of CUDC-907 were mediated by decreased NFκB/Forkhead box M1 (FOXM1) recruitment to promoters of genes involved in the DNA damage response; exogenous expression of NFκB/FOXM1 protected from CUDC-907-induced cytotoxicity. Together, these findings reveal CUDC-907 as a novel radiosensitizer with potent antitumor activity in pHGG and DIPG and provide a preclinical rationale for the combination of CUDC-907 with radiotherapy as a novel therapeutic strategy against pHGG and DIPG. More globally, we have identified NFκB and FOXM1 and their downstream transcriptional elements as critical targets for new treatments for pHGG and DIPG.Significance: These findings describe the radiosensitizing effect of a novel agent in pediatric high-grade gliomas, addressing a critical unmet need of increasing the radiation sensitivity of these highly aggressive tumors. Cancer Res; 78(14); 4007-21. ©2018 AACR.


Subject(s)
DNA Damage/drug effects , Forkhead Box Protein M1/metabolism , Glioma/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , NF-kappa B/metabolism , Phosphoinositide-3 Kinase Inhibitors , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Child , DNA Repair/drug effects , Glioma/metabolism , Homologous Recombination/drug effects , Humans , Mice , Mice, Nude , Morpholines/pharmacology , Pyrimidines/pharmacology , Radiation-Sensitizing Agents/pharmacology
4.
Cell Rep ; 18(13): 3167-3177, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28355568

ABSTRACT

During development of the vertebrate CNS, the basic helix-loop-helix (bHLH) transcription factor Olig2 sustains replication competence of progenitor cells that give rise to neurons and oligodendrocytes. A pathological counterpart of this developmental function is seen in human glioma, wherein Olig2 is required for maintenance of stem-like cells that drive tumor growth. The mitogenic/gliomagenic functions of Olig2 are regulated by phosphorylation of a triple serine motif (S10, S13, and S14) in the amino terminus. Here, we identify a set of three serine/threonine protein kinases (glycogen synthase kinase 3α/ß [GSK3α/ß], casein kinase 2 [CK2], and cyclin-dependent kinases 1/2 [CDK1/2]) that are, collectively, both necessary and sufficient to phosphorylate the triple serine motif. We show that phosphorylation of the motif itself serves as a template to prime phosphorylation of additional serines and creates a highly charged "acid blob" in the amino terminus of Olig2. Finally, we show that small molecule inhibitors of this forward-feeding phosphorylation cascade have potential as glioma therapeutics.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Glioma/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Animals , Casein Kinase II/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinases/metabolism , Disease Models, Animal , Glioma/pathology , Glycogen Synthase Kinase 3/metabolism , Humans , Mice , Phosphorylation/drug effects , Phosphoserine/metabolism , Small Molecule Libraries/pharmacology , Tumor Suppressor Protein p53/metabolism
5.
Elife ; 62017 02 08.
Article in English | MEDLINE | ID: mdl-28177281

ABSTRACT

Genomic instability is a hallmark of human cancer, and results in widespread somatic copy number alterations. We used a genome-scale shRNA viability screen in human cancer cell lines to systematically identify genes that are essential in the context of particular copy-number alterations (copy-number associated gene dependencies). The most enriched class of copy-number associated gene dependencies was CYCLOPS (Copy-number alterations Yielding Cancer Liabilities Owing to Partial losS) genes, and spliceosome components were the most prevalent. One of these, the pre-mRNA splicing factor SF3B1, is also frequently mutated in cancer. We validated SF3B1 as a CYCLOPS gene and found that human cancer cells harboring partial SF3B1 copy-loss lack a reservoir of SF3b complex that protects cells with normal SF3B1 copy number from cell death upon partial SF3B1 suppression. These data provide a catalog of copy-number associated gene dependencies and identify partial copy-loss of wild-type SF3B1 as a novel, non-driver cancer gene dependency.


Subject(s)
Gene Dosage , Neoplasms/genetics , Neoplasms/pathology , Phosphoproteins/genetics , RNA Splicing Factors/genetics , Cell Line, Tumor , Humans
6.
Neuro Oncol ; 19(6): 774-785, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28082416

ABSTRACT

Background: Activating mutations or structural rearrangements in BRAF are identified in roughly 75% of all pediatric low-grade astrocytomas (PLGAs). However, first-generation RAF inhibitors approved for adult melanoma have poor blood-brain penetrance and are only effective on tumors that express the canonical BRAFV600E oncoprotein, which functions as a monomer. These drugs (type I antagonists that target the "DFG-in" conformation of the kinase) fail to block signaling via KIAA1549:BRAF, a truncation/fusion BRAF oncoprotein which functions as a dimer and is found in the most common form of PLGA. Methods: A panel of small molecule RAF inhibitors (including type II inhibitors, targeting the "DFG-out" conformation of the kinase) was screened for drugs showing efficacy on murine models of PLGA and on authentic human PLGA cells expressing KIAA1549:BRAF. Results: We identify a type II RAF inhibitor that serves as an equipotent antagonist of BRAFV600E, KIAA1549:BRAF, and other noncanonical BRAF oncoproteins that function as dimers. This drug (MLN2480, also known as TAK-580) has good brain penetrance and is active on authentic human PLGA cells in brain organotypic cultures. Conclusion: MLN2480 may be an effective therapeutic for BRAF mutant pediatric astrocytomas.


Subject(s)
Astrocytoma/drug therapy , Brain Neoplasms/drug therapy , Heterocyclic Compounds, 3-Ring/pharmacology , Oncogene Proteins, Fusion/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Multimerization/drug effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , raf Kinases/antagonists & inhibitors , Animals , Astrocytoma/metabolism , Astrocytoma/pathology , Blood-Brain Barrier/drug effects , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Child , Heterocyclic Compounds, 3-Ring/chemistry , High-Throughput Screening Assays , Humans , Male , Mice , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , raf Kinases/genetics , raf Kinases/metabolism
7.
ACS Chem Biol ; 11(11): 3146-3153, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27643505

ABSTRACT

Basic helix-loop-helix (bHLH) transcription factors play critical roles in organism development and disease by regulating cell proliferation and differentiation. Transcriptional activity, whether by bHLH homo- or heterodimerization, is dependent on protein-protein and protein-DNA interactions mediated by α-helices. Thus, α-helical decoys have been proposed as potential targeted therapies for pathologic bHLH transcription. Here, we developed a library of stabilized α-helices of OLIG2 (SAH-OLIG2) to test the capacity of hydrocarbon-stapled peptides to disrupt OLIG2 homodimerization, which drives the development and chemoresistance of glioblastoma multiforme, one of the deadliest forms of human brain cancer. Although stapling successfully reinforced the α-helical structure of bHLH constructs of varying length, sequence-specific dissociation of OLIG2 dimers from DNA was not achieved. Re-evaluation of the binding determinants for OLIG2 self-association and stability revealed an unanticipated role of the C-terminal domain. These data highlight potential pitfalls in peptide-based targeting of bHLH transcription factors given the liabilities of their positively charged amino acid sequences and multifactorial binding determinants.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hydrocarbons/chemistry , Peptides/chemistry , Animals , COS Cells , Dimerization , Humans , Molecular Mimicry
8.
Cell Rep ; 16(4): 950-966, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27396340

ABSTRACT

In glioblastoma, invasion and proliferation are presumed to be mutually exclusive events; however, the molecular mechanisms that mediate this switch at the cellular level remain elusive. Previously, we have shown that phospho-OLIG2, a central-nervous-system-specific transcription factor, is essential for tumor growth and proliferation. Here, we show that the modulation of OLIG2 phosphorylation can trigger a switch between proliferation and invasion. Glioma cells with unphosphorylated OLIG2(S10, S13, S14) are highly migratory and invasive, both in vitro and in vivo. Mechanistically, unphosphorylated OLIG2 induces TGF-ß2 expression and promotes invasive mesenchymal properties in glioma cells. Inhibition of the TGF-ß2 pathway blocks this OLIG2-dependent invasion. Furthermore, ectopic expression of phosphomimetic Olig2 is sufficient to block TGF-ß2-mediated invasion and reduce expression of invasion genes (ZEB1 and CD44). Our results not only provide a mechanistic insight into how cells switch from proliferation to invasion but also offer therapeutic opportunities for inhibiting dissemination of gliomas.


Subject(s)
Glioblastoma/genetics , Glioblastoma/pathology , Neoplasm Invasiveness/genetics , Oligodendrocyte Transcription Factor 2/genetics , Protein Processing, Post-Translational/genetics , Transforming Growth Factor beta/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Humans , Hyaluronan Receptors/genetics , Mice , Mice, Nude , Neoplasm Invasiveness/pathology , Phosphorylation/genetics , Signal Transduction/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics
9.
J Neurosci ; 34(25): 8507-18, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24948806

ABSTRACT

The bHLH transcription factor Olig2 is expressed in cycling neural progenitor cells but also in terminally differentiated, myelinating oligodendrocytes. Sustained expression of Olig2 is counterintuitive because all known functions of the protein in expansion of neural progenitors and specification of oligodendrocyte progenitors are completed with the formation of mature white matter. How are the biological functions of Olig2 suppressed in terminally differentiated oligodendrocytes? In previous studies, we have shown that a triple serine motif in the amino terminus of Olig2 is phosphorylated in cycling neural progenitors but not in their differentiated progeny. We now show that phosphorylation of the triple serine motif regulates intranuclear compartmentalization of murine Olig2. Phosphorylated Olig2 is preferentially localized to a transcriptionally active "open" chromatin compartment together with coregulator proteins essential for regulation of gene expression. Unphosphorylated Olig2, as seen in mature white matter, is localized mainly within a transcriptionally inactive, chromatin fraction characterized by condensed and inaccessible DNA. Of special note is the observation that the p53 tumor suppressor protein is confined to the open chromatin fraction. Proximity ligation assays show that phosphorylation brings Olig2 within 30 nm of p53 within the open chromatin compartment. The data thus shed light on previously noted promitogenic functions of phosphorylated Olig2, which reflect, at least in part, an oppositional relationship with p53 functions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Neural Stem Cells/chemistry , Neural Stem Cells/metabolism , Amino Acid Motifs/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Nucleus/genetics , Cells, Cultured , Female , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Oligodendrocyte Transcription Factor 2 , Phosphorylation/genetics , Pregnancy
10.
Neuron ; 81(3): 574-87, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24507192

ABSTRACT

Abnormal GABAergic interneuron density, and imbalance of excitatory versus inhibitory tone, is thought to result in epilepsy, neurodevelopmental disorders, and psychiatric disease. Recent studies indicate that interneuron cortical density is determined primarily by the size of the precursor pool in the embryonic telencephalon. However, factors essential for regulating interneuron allocation from telencephalic multipotent precursors are poorly understood. Here we report that Olig1 represses production of GABAergic interneurons throughout the mouse brain. Olig1 deletion in mutant mice results in ectopic expression and upregulation of Dlx1/2 genes in the ventral medial ganglionic eminences and adjacent regions of the septum, resulting in an ∼30% increase in adult cortical interneuron numbers. We show that Olig1 directly represses the Dlx1/2 I12b intergenic enhancer and that Dlx1/2 functions genetically downstream of Olig1. These findings establish Olig1 as an essential repressor of Dlx1/2 and interneuron production in developing mammalian brain.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/cytology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , Interneurons/physiology , Transcription Factors/metabolism , Action Potentials/genetics , Action Potentials/physiology , Age Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain/embryology , Brain/growth & development , Cell Count , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Glutamate Decarboxylase/metabolism , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Organ Culture Techniques , Patch-Clamp Techniques , Synapses/physiology , Transcription Factors/genetics
11.
Sci Rep ; 3: 2859, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-24091529

ABSTRACT

Drug transit through the blood-brain barrier (BBB) is essential for therapeutic responses in malignant glioma. Conventional methods for assessment of BBB penetrance require synthesis of isotopically labeled drug derivatives. Here, we report a new methodology using matrix assisted laser desorption ionization mass spectrometry imaging (MALDI MSI) to visualize drug penetration in brain tissue without molecular labeling. In studies summarized here, we first validate heme as a simple and robust MALDI MSI marker for the lumen of blood vessels in the brain. We go on to provide three examples of how MALDI MSI can provide chemical and biological insights into BBB penetrance and metabolism of small molecule signal transduction inhibitors in the brain - insights that would be difficult or impossible to extract by use of radiolabeled compounds.


Subject(s)
Blood-Brain Barrier/metabolism , Molecular Imaging/methods , Pharmaceutical Preparations/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Biomarkers/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Disease Models, Animal , Erlotinib Hydrochloride , Glioma/metabolism , Glioma/pathology , Heme/metabolism , Heterografts , Humans , Mice , Neovascularization, Pathologic , Optical Imaging/methods , Permeability , Pharmaceutical Preparations/chemistry , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/chemistry , Quinazolines/metabolism , Quinazolines/pharmacokinetics , Reproducibility of Results
12.
Blood ; 119(21): 4878-88, 2012 May 24.
Article in English | MEDLINE | ID: mdl-22474248

ABSTRACT

The transcription factor C/EBPα is a critical mediator of myeloid differentiation and is often functionally impaired in acute myeloid leukemia. Recent studies have suggested that oncogenic FLT3 activity disrupts wild-type C/EBPα function via phosphorylation on serine 21 (S21). Despite the apparent role of pS21 as a negative regulator of C/EBPα transcription activity, the mechanism by which phosphorylation tips the balance between transcriptionally competent and inhibited forms remains unresolved. In the present study, we used immuno-affinity purification combined with quantitative mass spectrometry to delineate the proteins associated with C/EBPα on chromatin. We identified DEK, a protein with genetic links to leukemia, as a member of the C/EBPα complexes, and demonstrate that this association is disrupted by S21 phosphorylation. We confirmed that DEK is recruited specifically to chromatin with C/EBPα to enhance GCSFR3 promoter activation. In addition, we demonstrated that genetic depletion of DEK reduces the ability of C/EBPα to drive the expression of granulocytic target genes in vitro and disrupts G-CSF-mediated granulocytic differentiation of fresh human BM-derived CD34(+) cells. Our data suggest that C/EBPα and DEK coordinately activate myeloid gene expression and that S21 phosphorylation on wild-type C/EBPα mediates protein interactions that regulate the differentiation capacity of hematopoietic progenitors.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Cell Differentiation/genetics , Chromosomal Proteins, Non-Histone/physiology , Myeloid Cells/physiology , Oncogene Proteins/physiology , Antibodies/pharmacology , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , HEK293 Cells , Hematopoiesis/drug effects , Hematopoiesis/genetics , Humans , K562 Cells , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Poly-ADP-Ribose Binding Proteins , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology
13.
Cancer Cell ; 19(3): 359-71, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21397859

ABSTRACT

High-grade gliomas are notoriously insensitive to radiation and genotoxic drugs. Paradoxically, the p53 gene is structurally intact in the majority of these tumors. Resistance to genotoxic modalities in p53-positive gliomas is generally attributed to attenuation of p53 functions by mutations of other components within the p53 signaling axis, such as p14(Arf), MDM2, and ATM, but this explanation is not entirely satisfactory. We show here that the central nervous system (CNS)-restricted transcription factor Olig2 affects a key posttranslational modification of p53 in both normal and malignant neural progenitors and thereby antagonizes the interaction of p53 with promoter elements of multiple target genes. In the absence of Olig2 function, even attenuated levels of p53 are adequate for biological responses to genotoxic damage.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA Damage , Glioma/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/radiation effects , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Survival/radiation effects , Cells, Cultured , Central Nervous System/metabolism , Central Nervous System/pathology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Flow Cytometry , Glioma/genetics , Glioma/pathology , HEK293 Cells , Humans , Immunoblotting , Male , Mice , Mice, 129 Strain , Mice, Knockout , Mice, SCID , Nerve Tissue Proteins/genetics , Neural Stem Cells/radiation effects , Oligodendrocyte Transcription Factor 2 , Phosphorylation , Promoter Regions, Genetic/genetics , Protein Binding , Tumor Suppressor Protein p53/genetics
14.
Neuron ; 69(5): 906-17, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21382551

ABSTRACT

The bHLH transcription factors that regulate early development of the central nervous system can generally be classified as either antineural or proneural. Initial expression of antineural factors prevents cell cycle exit and thereby expands the pool of neural progenitors. Subsequent (and typically transient) expression of proneural factors promotes cell cycle exit, subtype specification, and differentiation. Against this backdrop, the bHLH transcription factor Olig2 in the oligodendrocyte lineage is unorthodox, showing antineural functions in multipotent CNS progenitor cells but also sustained expression and proneural functions in the formation of oligodendrocytes. We show here that the proliferative function of Olig2 is controlled by developmentally regulated phosphorylation of a conserved triple serine motif within the amino-terminal domain. In the phosphorylated state, Olig2 maintains antineural (i.e., promitotic) functions that are reflected in human glioma cells and in a genetically defined murine model of primary glioma.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Phosphorylation/physiology , Analysis of Variance , Animals , Blotting, Western , Cell Lineage/physiology , Chromatin Immunoprecipitation , Humans , Mice , Oligodendrocyte Transcription Factor 2 , Reverse Transcriptase Polymerase Chain Reaction
15.
Cancer Cell ; 11(5): 395-406, 2007 May.
Article in English | MEDLINE | ID: mdl-17482130

ABSTRACT

PPARgamma is a member of the nuclear receptor family for which agonist ligands have antigrowth effects. However, clinical studies using PPARgamma ligands as a monotherapy failed to show a beneficial effect. Here we have studied the effects of PPARgamma activation with chemotherapeutic agents in current use for specific cancers. We observed a striking synergy between rosiglitazone and platinum-based drugs in several different cancers both in vitro and using transplantable and chemically induced "spontaneous" tumor models. The effect appears to be due in part to PPARgamma-mediated downregulation of metallothioneins, proteins that have been shown to be involved in resistance to platinum-based therapy. These data strongly suggest combining PPARgamma agonists and platinum-based drugs for the treatment of certain human cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Carboplatin/pharmacology , Cell Division/drug effects , PPAR gamma/agonists , Thiazolidinediones/pharmacology , Drug Synergism , Ligands , Rosiglitazone
16.
Neuron ; 53(4): 503-17, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17296553

ABSTRACT

Recent studies have identified stem cells in brain cancer. However, their relationship to normal CNS progenitors, including dependence on common lineage-restricted pathways, is unclear. We observe expression of the CNS-restricted transcription factor, OLIG2, in human glioma stem and progenitor cells reminiscent of type C transit-amplifying cells in germinal zones of the adult brain. Olig2 function is required for proliferation of neural progenitors and for glioma formation in a genetically relevant murine model. Moreover, we show p21(WAF1/CIP1), a tumor suppressor and inhibitor of stem cell proliferation, is directly repressed by OLIG2 in neural progenitors and gliomas. Our findings identify an Olig2-regulated lineage-restricted pathway critical for proliferation of normal and tumorigenic CNS stem cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/pathology , Glioma/pathology , Nerve Tissue Proteins/metabolism , Neurons/physiology , Stem Cells/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Bromodeoxyuridine/metabolism , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation/methods , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Embryo, Mammalian , Flow Cytometry , Green Fluorescent Proteins/biosynthesis , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Nerve Tissue Proteins/deficiency , Oligodendrocyte Transcription Factor 2
17.
Proc Natl Acad Sci U S A ; 103(20): 7853-8, 2006 May 16.
Article in English | MEDLINE | ID: mdl-16682644

ABSTRACT

In the adult central nervous system, two distinct populations of glial cells expressing the chondroitin sulfate proteoglycan NG2 have been described: bipolar progenitor cells and more differentiated "synantocytes." These cells have diverse neurological functions, including critical roles in synaptic transmission, repair, and regeneration. Despite their potential importance, the genetic factors that regulate NG2 cell development are poorly understood, and the relationship of synantocytes to the oligodendroglial lineage, in particular, remains controversial. Here, we show that >90% of embryonic and adult NG2 cells express Olig2, a basic helix-loop-helix transcription factor required for oligodendrocyte lineage specification. Analysis of mice lacking Olig function demonstrates a failure of NG2 cell development at embryonic and perinatal stages that can be rescued by addition of a transgene containing the human OLIG2 locus. These findings show a general requirement for Olig function in NG2 cell development and highlight further roles for Olig transcription factors in neural progenitor cells.


Subject(s)
Antigens/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/embryology , Nerve Tissue Proteins/metabolism , Neurons/physiology , Oligodendroglia/physiology , Proteoglycans/metabolism , Stem Cells/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain/anatomy & histology , Brain/metabolism , Cell Lineage , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/cytology , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/cytology , Spinal Cord/cytology , Spinal Cord/embryology , Spinal Cord/metabolism , Stem Cells/cytology
18.
J Neurosci ; 25(13): 3478-87, 2005 Mar 30.
Article in English | MEDLINE | ID: mdl-15800203

ABSTRACT

How do myelinated axons signal to the nuclei of cells that enwrap them? The cell bodies of oligodendrocytes and Schwann cells are segregated from axons by multiple layers of bimolecular lipid leaflet and myelin proteins. Conventional signal transduction strategies would seem inadequate to the challenge without special adaptations. Wallerian degeneration provides a model to study axon-to-Schwann cell signaling in the context of nerve injury. We show a hitherto undetected rapid, but transient, activation of the receptor tyrosine kinase erbB2 in myelinating Schwann cells after sciatic nerve axotomy. Deconvolving microscopy using phosphorylation state-specific antibodies shows that erbB2 activation emanates from within the microvilli of Schwann cells, in direct contact with the axons they enwrap. To define the functional role of this transient activation, we used a small molecule antagonist of erbB2 activation (PKI166). The response of myelinating Schwann cells to axotomy is inhibited by PKI166 in vivo. Using neuron/Schwann cell cocultures prepared in compartmentalized cell culture chambers, we show that even transient activation of erbB2 is sufficient to initiate Schwann cell demyelination and that the initiating functions of erbB2 are localized to Schwann cells.


Subject(s)
Neuroglia/physiology , Neurons/physiology , Signal Transduction/physiology , Wallerian Degeneration/pathology , Analysis of Variance , Animals , Axotomy/methods , Blotting, Western/methods , Bromodeoxyuridine/metabolism , Cell Proliferation , Cells, Cultured , Coculture Techniques/methods , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Embryo, Mammalian , Female , Fluorescent Antibody Technique/methods , Ganglia, Spinal/pathology , Gene Expression/drug effects , Gene Expression/physiology , Glycoproteins/metabolism , Immunoprecipitation/methods , Mitogen-Activated Protein Kinase Kinases/metabolism , Myelin Basic Protein/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , Neuregulins/pharmacology , Neuroglia/drug effects , Neurons/drug effects , Platelet-Derived Growth Factor/pharmacology , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-2 , Schwann Cells/pathology , Sciatic Neuropathy/pathology , Signal Transduction/drug effects , Sodium Channels/metabolism , Time Factors , Wallerian Degeneration/physiopathology
19.
J Neurobiol ; 62(1): 121-33, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15389679

ABSTRACT

Cortical progenitor cells from rat embryos give rise to neurons or glia following exposure to platelet derived growth factor (PDGF) or ciliary neurotrophic factor (CNTF), respectively. Both growth factors impart their developmental cues quickly through a transcription-dependent mechanism. Do the alternate developmental responses to PDGF and CNTF reflect induction of qualitatively distinct genes? Alternatively, do the same genes respond to each growth factor, but with quantitatively distinct kinetics? Using differential library screening and custom cDNA microarrays we show that a common set of genes responds to either growth factor. However, quantitative differences in the onset and duration of gene induction equate to the expression of factor-specific gene signatures. Multitissue cluster analysis also reveals tissue-specific gene signatures that may play important roles in the developing brain.


Subject(s)
Cell Differentiation/genetics , Cerebral Cortex/embryology , Gene Expression Regulation, Developmental/genetics , Neuroglia/metabolism , Neurons/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Cerebral Cortex/anatomy & histology , Cerebral Cortex/metabolism , Ciliary Neurotrophic Factor/metabolism , Ciliary Neurotrophic Factor/pharmacology , Cluster Analysis , Cues , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation, Developmental/drug effects , Gene Library , Oligonucleotide Array Sequence Analysis , Platelet-Derived Growth Factor/metabolism , Platelet-Derived Growth Factor/pharmacology , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects , Transcriptional Activation
20.
Science ; 306(5705): 2255-7, 2004 Dec 24.
Article in English | MEDLINE | ID: mdl-15618518

ABSTRACT

In the developing brain, transcription factors (TFs) direct the formation of a diverse array of neurons and glia. We identifed 1445 putative TFs in the mouse genome. We used in situ hybridization to map the expression of over 1000 of these TFs and TF-coregulator genes in the brains of developing mice. We found that 349 of these genes showed restricted expression patterns that were adequate to describe the anatomical organization of the brain. We provide a comprehensive inventory of murine TFs and their expression patterns in a searchable brain atlas database.


Subject(s)
Brain/growth & development , Brain/metabolism , Gene Expression Profiling , Genome , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Animals, Newborn , Brain/anatomy & histology , Brain/embryology , Cloning, Molecular , Corpus Striatum/anatomy & histology , Corpus Striatum/embryology , Corpus Striatum/growth & development , Corpus Striatum/metabolism , DNA Primers , Databases, Factual , Hypothalamus/anatomy & histology , Hypothalamus/embryology , Hypothalamus/growth & development , Hypothalamus/metabolism , In Situ Hybridization , Mesencephalon/anatomy & histology , Mesencephalon/embryology , Mesencephalon/growth & development , Mesencephalon/metabolism , Mice , Neocortex/anatomy & histology , Neocortex/embryology , Neocortex/growth & development , Neocortex/metabolism , Polymerase Chain Reaction , Rhombencephalon/anatomy & histology , Rhombencephalon/embryology , Rhombencephalon/growth & development , Rhombencephalon/metabolism , Spinal Cord/anatomy & histology , Spinal Cord/embryology , Spinal Cord/growth & development , Spinal Cord/metabolism , Thalamus/anatomy & histology , Thalamus/embryology , Thalamus/growth & development , Thalamus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...