Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Rep ; 9(3): 1034-46, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25437558

ABSTRACT

The development of targeted therapeutics for neuroblastoma, the third most common tumor in children, has been limited by a poor understanding of growth signaling mechanisms unique to the peripheral nerve precursors from which tumors arise. In this study, we combined genetics with gene-expression analysis in the peripheral sympathetic nervous system to implicate arginase 1 and GABA signaling in tumor formation in vivo. In human neuroblastoma cells, either blockade of ARG1 or benzodiazepine-mediated activation of GABA-A receptors induced apoptosis and inhibited mitogenic signaling through AKT and MAPK. These results suggest that ARG1 and GABA influence both neural development and neuroblastoma and that benzodiazepines in clinical use may have potential applications for neuroblastoma therapy.


Subject(s)
Arginase/genetics , Brain Neoplasms/genetics , Molecular Targeted Therapy , Neuroblastoma/genetics , Quantitative Trait Loci/genetics , Receptors, GABA-A/genetics , Animals , Apoptosis , Arginase/antagonists & inhibitors , Cell Line, Tumor , Cell Survival , Chromosomes, Mammalian/genetics , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genetic Linkage , Genetic Predisposition to Disease , Humans , Mice , Survival Analysis , gamma-Aminobutyric Acid/metabolism
2.
Cancer Cell ; 18(6): 669-82, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21156288

ABSTRACT

Malignant astrocytic brain tumors are among the most lethal cancers. Quiescent and therapy-resistant neural stem cell (NSC)-like cells in astrocytomas are likely to contribute to poor outcome. Malignant oligodendroglial brain tumors, in contrast, are therapy sensitive. Using magnetic resonance imaging (MRI) and detailed developmental analyses, we demonstrated that murine oligodendroglioma cells show characteristics of oligodendrocyte progenitor cells (OPCs) and are therapy sensitive, and that OPC rather than NSC markers enriched for tumor formation. MRI of human oligodendroglioma also suggested a white matter (WM) origin, with markers for OPCs rather than NSCs similarly enriching for tumor formation. Our results suggest that oligodendroglioma cells show hallmarks of OPCs, and that a progenitor rather than a NSC origin underlies improved prognosis in patients with this tumor.


Subject(s)
Brain Neoplasms/pathology , Neural Stem Cells/pathology , Oligodendroglia/pathology , Oligodendroglioma/pathology , Animals , Antigens/analysis , Benzamides/pharmacology , Cell Differentiation , Cell Line, Tumor , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Humans , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Oncogene Proteins v-erbB/analysis , Proteoglycans/analysis , Temozolomide , Tumor Suppressor Protein p53/physiology
3.
Genes Dev ; 24(10): 1059-72, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20478998

ABSTRACT

Medulloblastoma (MB) is the most common malignant brain tumor of childhood. Sonic Hedgehog (SHH) signaling drives a minority of MB, correlating with desmoplastic pathology and favorable outcome. The majority, however, arises independently of SHH and displays classic or large cell anaplastic (LCA) pathology and poor prognosis. To identify common signaling abnormalities, we profiled mRNA, demonstrating misexpression of MYCN in the majority of human MB and negligible expression in normal cerebella. We clarified a role in pathogenesis by targeting MYCN (and luciferase) to cerebella of transgenic mice. MYCN-driven MB showed either classic or LCA pathologies, with Shh signaling activated in approximately 5% of tumors, demonstrating that MYCN can drive MB independently of Shh. MB arose at high penetrance, consistent with a role for MYCN in initiation. Tumor burden correlated with bioluminescence, with rare metastatic spread to the leptomeninges, suggesting roles for MYCN in both progression and metastasis. Transient pharmacological down-regulation of MYCN led to both clearance and senescence of tumor cells, and improved survival. Targeted expression of MYCN thus contributes to initiation, progression, and maintenance of MB, suggesting a central role for MYCN in pathogenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Medulloblastoma/physiopathology , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Animals , Cell Cycle/physiology , Cellular Senescence/physiology , Cerebellum/metabolism , Down-Regulation , Gene Expression Profiling , Genomic Instability , Hedgehog Proteins/metabolism , Humans , Medulloblastoma/pathology , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neoplasm Metastasis/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics
4.
Neoplasia ; 10(11): 1268-74, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18953436

ABSTRACT

Chemoresistance in neuroblastoma is a significant issue complicating treatment of this common pediatric solid tumor. MYCN-amplified neuroblastomas are infrequently mutated at p53 and are chemosensitive at diagnosis but acquire p53 mutations and chemoresistance with relapse. Paradoxically, Myc-driven transformation is thought to require apoptotic blockade. We used the TH-MYCN transgenic murine model to examine the role of p53-driven apoptosis on neuroblastoma tumorigenesis and the response to chemotherapy. Tumors formed with high penetrance and low latency in p53-haploinsufficient TH-MYCN mice. Cyclophosphamide (CPM) induced a complete remission in p53 wild type TH-MYCN tumors, mirroring the sensitivity of childhood neuroblastoma to this agent. Treated tumors showed a prominent proliferation block, induction of p53 protein, and massive apoptosis proceeding through induction of the Bcl-2 homology domain-3-only proteins PUMA and Bim, leading to the activation of Bax and cleavage of caspase-3 and -9. Apoptosis induced by CPM was reduced in p53-haploinsufficient tumors. Treatment of MYCN-expressing human neuroblastoma cell lines with CPM induced apoptosis that was suppressible by siRNA to p53. Taken together, the results indicate that the p53 pathway plays a significant role in opposing MYCN-driven oncogenesis in a mouse model of neuroblastoma and that basal inactivation of the pathway is achieved in progressing tumors. This, in part, explains the striking sensitivity of such tumors to chemotoxic agents that induce p53-dependent apoptosis and is consistent with clinical observations that therapy-associated mutations in p53 are a likely contributor to the biology of tumors at relapse and secondarily mediate resistance to therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Cyclophosphamide/therapeutic use , Genes, p53 , Neuroblastoma/drug therapy , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Proliferation , Child , Cyclophosphamide/pharmacology , Heterozygote , Humans , Mice , Mice, Transgenic , Mutation , N-Myc Proto-Oncogene Protein , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Remission Induction
5.
Cancer Res ; 67(19): 9435-42, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17909053

ABSTRACT

Targeted expression of MYCN to the neural crest [under control of the rat tyrosine hydroxylase (TH) promoter] causes neuroblastoma in transgenic mice (TH-MYCN) and is a well-established model for this disease. Because high levels of MYCN are associated with enhanced tumor angiogenesis and poor clinical outcome in neuroblastoma, we serially characterized malignant progression, angiogenesis, and sensitivity to angiogenic blockade in tumors from these animals. Tumor cells were proliferative, secreted high levels of the angiogenic ligand vascular endothelial growth factor (VEGF), and recruited a complex vasculature expressing the angiogenic markers VEGF-R2, alpha-SMA, and matrix metalloproteinases MMP-2 and MMP-9, all of which are also expressed in human disease. Treatment of established murine tumors with the angiogenesis inhibitor TNP-470 caused near-complete ablation, with reduced proliferation, enhanced apoptosis, and vasculature disruption. Because TNP-470 has been associated with neurotoxicity, we tested the recently described water-soluble HPMA copolymer-TNP-470 conjugate (caplostatin), which showed comparable efficacy and was well tolerated without weight loss or neurotoxicity as measured by rotarod testing. This study highlights the importance of angiogenesis inhibition in a spontaneous murine tumor with native tumor-microenvironment interactions, validates the use of mice transgenic for TH-MYCN as a model for therapy in this common pediatric tumor, and supports further clinical development of caplostatin as an antiangiogenic therapy in childhood neuroblastoma.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cyclohexanes/pharmacology , Neuroblastoma/blood supply , Neuroblastoma/drug therapy , Sesquiterpenes/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Disease Models, Animal , Disease Progression , Genes, myc , Mice , Mice, Transgenic , Neovascularization, Pathologic/drug therapy , Neuroblastoma/pathology , O-(Chloroacetylcarbamoyl)fumagillol , Rats
6.
J Cell Biochem ; 102(6): 1529-41, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17486636

ABSTRACT

Neuroblastoma is a common pediatric malignancy that metastasizes to the liver, bone, and other organs. Children with metastatic disease have a less than 50% chance of survival with current treatments. Insulin-like growth factors (IGFs) stimulate neuroblastoma growth, survival, and motility, and are expressed by neuroblastoma cells and the tissues they invade. Thus, therapies that disrupt the effects of IGFs on neuroblastoma tumorigenesis may slow disease progression. We show that NVP-AEW541, a specific inhibitor of the IGF-I receptor (IGF-IR), potently inhibits neuroblastoma growth in vitro. Nordihydroguaiaretic acid (NDGA), a phenolic compound isolated from the creosote bush (Larrea divaricata), has anti-tumor properties against a number of malignancies, has been shown to inhibit the phosphorylation and activation of the IGF-IR in breast cancer cells, and is currently in Phase I trials for prostate cancer. In the present study in neuroblastoma, NDGA inhibits IGF-I-mediated activation of the IGF-IR and disrupts activation of ERK and Akt signaling pathways induced by IGF-I. NDGA inhibits growth of neuroblastoma cells and induces apoptosis at higher doses, causing IGF-I-resistant activation of caspase-3 and a large increase in the fraction of sub-G0 cells. In addition, NDGA inhibits the growth of xenografted human neuroblastoma tumors in nude mice. These results indicate that NDGA may be useful in the treatment of neuroblastoma and may function in part via disruption of IGF-IR signaling.


Subject(s)
Cell Proliferation/drug effects , Insulin-Like Growth Factor I/antagonists & inhibitors , Masoprocol/pharmacology , Neuroblastoma/drug therapy , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Culture Techniques , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mice , Mice, Nude , Neuroblastoma/pathology , Phosphorylation/drug effects , Propidium/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Xenograft Model Antitumor Assays
7.
Cancer Res ; 66(16): 8139-46, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16912192

ABSTRACT

Amplification of MYCN occurs commonly in neuroblastoma. We report that phosphatidylinositol 3-kinase (PI3K) inhibition in murine neuroblastoma (driven by a tyrosine hydroxylase-MYCN transgene) led to decreased tumor mass and decreased levels of Mycn protein without affecting levels of MYCN mRNA. Consistent with these observations, PI3K inhibition in MYCN-amplified human neuroblastoma cell lines resulted in decreased levels of Mycn protein without affecting levels of MYCN mRNA and caused decreased proliferation and increased apoptosis. To clarify the importance of Mycn as a target of broad-spectrum PI3K inhibitors, we transduced wild-type N-myc and N-myc mutants lacking glycogen synthase kinase 3beta phosphorylation sites into human neuroblastoma cells with no endogenous expression of myc. In contrast to wild-type N-myc, the phosphorylation-defective mutant proteins were stabilized and were resistant to the antiproliferative effects of PI3K inhibition. Our results show the importance of Mycn as a therapeutic target in established tumors in vivo, offer a mechanistic rationale to test PI3K inhibitors in MYCN-amplified neuroblastoma, and represent a therapeutic approach applicable to a broad range of cancers in which transcription factors are stabilized through a PI3K-dependent mechanism.


Subject(s)
Neuroblastoma/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Phosphoinositide-3 Kinase Inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Division , Cell Line, Tumor , Chromones/pharmacology , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Gene Amplification , Humans , Morpholines/pharmacology , N-Myc Proto-Oncogene Protein , Neuroblastoma/genetics
8.
Cancer Cell ; 9(5): 341-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16697955

ABSTRACT

The PI3 kinase family of lipid kinases promotes cell growth and survival by generating the second messenger phosphatidylinositol-3,4,5-trisphosphate. To define targets critical for cancers driven by activation of PI3 kinase, we screened a panel of potent and structurally diverse drug-like molecules that target this enzyme family. Surprisingly, a single agent (PI-103) effected proliferative arrest in glioma cells, despite the ability of many compounds to block PI3 kinase signaling through its downstream effector, Akt. The unique cellular activity of PI-103 was traced directly to its ability to inhibit both PI3 kinase alpha and mTOR. PI-103 showed significant activity in xenografted tumors with no observable toxicity. These data demonstrate an emergent efficacy due to combinatorial inhibition of mTOR and PI3 kinase alpha in malignant glioma.


Subject(s)
Glioma/drug therapy , Glioma/enzymology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinases/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Class I Phosphatidylinositol 3-Kinases , Enzyme Activation , ErbB Receptors/metabolism , Glioma/pathology , Humans , Mice , Mice, Inbred BALB C , Organoplatinum Compounds/pharmacology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction , Substrate Specificity , TOR Serine-Threonine Kinases , Treatment Outcome , Tumor Suppressor Protein p53/metabolism
9.
Cell ; 125(4): 733-47, 2006 May 19.
Article in English | MEDLINE | ID: mdl-16647110

ABSTRACT

Phosphoinositide 3-kinases (PI3-Ks) are an important emerging class of drug targets, but the unique roles of PI3-K isoforms remain poorly defined. We describe here an approach to pharmacologically interrogate the PI3-K family. A chemically diverse panel of PI3-K inhibitors was synthesized, and their target selectivity was biochemically enumerated, revealing cryptic homologies across targets and chemotypes. Crystal structures of three inhibitors bound to p110gamma identify a conformationally mobile region that is uniquely exploited by selective compounds. This chemical array was then used to define the PI3-K isoforms required for insulin signaling. We find that p110alpha is the primary insulin-responsive PI3-K in cultured cells, whereas p110beta is dispensable but sets a phenotypic threshold for p110alpha activity. Compounds targeting p110alpha block the acute effects of insulin treatment in vivo, whereas a p110beta inhibitor has no effect. These results illustrate systematic target validation using a matrix of inhibitors that span a protein family.


Subject(s)
Enzyme Inhibitors/chemistry , Insulin/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction/physiology , Adipose Tissue/cytology , Animals , Binding Sites , Class I Phosphatidylinositol 3-Kinases , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Female , Glucose/metabolism , Humans , Insulin Receptor Substrate Proteins , Isoenzymes/chemistry , Isoenzymes/genetics , Mice , Models, Molecular , Molecular Structure , Muscle Fibers, Skeletal/metabolism , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , Phosphoproteins/metabolism
10.
Cancer Res ; 65(9): 3617-23, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15867356

ABSTRACT

Tumors arise in part from the deleterious effects of genetic and epigenetic mechanisms on gene expression. In several mouse models of human tumors, the tumorigenic phenotype is reversible, suggesting that epigenetic mechanisms also contribute significantly to tumorigenesis in mice. It is not known whether these are the same epigenetic mechanisms in human and mouse tumors or whether they affect homologous genes. Using an integrated approach for genome-wide methylation and copy number analyses, we identified SLC5A8 on chromosome 12q23.1 that was affected frequently by aberrant methylation in human astrocytomas and oligodendrogliomas. SLC5A8 encodes a sodium monocarboxylate cotransporter that was highly expressed in normal brain but was significant down-regulated in primary gliomas. Bisulfite sequencing analysis showed that the CpG island was unmethylated in normal brain but frequently localized methylated in brain tumors, consistent with the tumor-specific loss of gene expression. In glioma cell lines, SLC5A8 expression was also suppressed but could be reactivated with a methylation inhibitor. Expression of exogenous SLC5A8 in LN229 and LN443 glioma cells inhibited colony formation, suggesting that it may function as a growth suppressor in normal brain cells. Remarkably, 9 of 10 murine oligodendroglial tumors (from p53+/- or ink4a/arf+/- animals transgenic for S100beta-v-erbB) showed a similar tumor-specific down-regulation of mSLC5A8, the highly conserved mouse homologue. Taken together, these data suggest that SLC5A8 functions as a growth suppressor gene in vitro and that it is silenced frequently by epigenetic mechanisms in primary gliomas. The shared epigenetic inactivation of mSLC5A8 in mouse gliomas indicates an additional degree of commonality in the origin and/or pathway to tumorigenesis between primary human tumors and these mouse models of gliomas.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Cation Transport Proteins/genetics , Gene Silencing , Genes, Tumor Suppressor , Oligodendroglioma/genetics , Animals , Astrocytoma/metabolism , Brain Neoplasms/metabolism , Cation Transport Proteins/biosynthesis , Cell Line, Tumor , CpG Islands/genetics , DNA Methylation , Disease Models, Animal , Down-Regulation , Gene Expression Regulation, Neoplastic , Genetic Vectors/genetics , Humans , Mice , Mice, Inbred DBA , Mice, Transgenic , Monocarboxylic Acid Transporters , Oligodendroglioma/metabolism , Retroviridae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...