Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Lett ; 377(1): 55-64, 2016 07 10.
Article in English | MEDLINE | ID: mdl-27102002

ABSTRACT

Glioblastoma (GBM) contains a population of stem-like cells that promote tumor invasion and resistance to therapy. Identifying and targeting stem cell factors in GBM may lead to the development of more effective therapies. High Mobility Group AT-hook 2 (HMGA2) is a transcriptional modulator that mediates motility and self-renewal in normal and cancer stem cells. We identified increased expression of HMGA2 in the majority of primary human GBM tumors and cell lines compared to normal brain. Additionally, HMGA2 expression was increased in CD133+ GBM neurosphere cells compared to CD133- cells. Targeting HMGA2 with lentiviral short hairpin RNA (shRNA) led to decreased GBM stemness, invasion, and tumorigenicity. Ectopic expression of HMGA2 in GBM cell lines promoted stemness, invasion, and tumorigenicity. Our data suggests that targeting HMGA2 in GBM may be therapeutically beneficial.


Subject(s)
Cell Movement , Cell Proliferation , Glioblastoma/metabolism , HMGA2 Protein/metabolism , Neoplastic Stem Cells/metabolism , AC133 Antigen/metabolism , Apoptosis , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , HMGA2 Protein/genetics , Humans , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Phenotype , RNA Interference , Signal Transduction , Spheroids, Cellular , Time Factors , Transfection , Tumor Burden , Up-Regulation , Xenograft Model Antitumor Assays
3.
J Neuropathol Exp Neurol ; 74(8): 778-90, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26115193

ABSTRACT

NOTCH regulates stem cells during normal development and stemlike cells in cancer, but the roles of NOTCH in the lethal pediatric brain tumor diffuse intrinsic pontine glioma (DIPG) remain unknown. Because DIPGs express stem cell factors such as SOX2 and MYCN, we hypothesized that NOTCH activity would be critical for DIPG growth. We determined that primary DIPGs expressed high levels of NOTCH receptors, ligands, and downstream effectors. Treatment of the DIPG cell lines JHH-DIPG1 and SF7761 with the γ-secretase inhibitor MRK003 suppressed the level of the NOTCH effectors HES1, HES4, and HES5; inhibited DIPG growth by 75%; and caused a 3-fold induction of apoptosis. Short hairpin RNAs targeting the canonical NOTCH pathway caused similar effects. Pretreatment of DIPG cells with MRK003 suppressed clonogenic growth by more than 90% and enhanced the efficacy of radiation therapy. The high level of MYCN in DIPG led us to test sequential therapy with the bromodomain inhibitor JQ1 and MRK003, and we found that JQ1 and MRK003 inhibited DIPG growth and induced apoptosis. Together, these results suggest that dual targeting of NOTCH and MYCN in DIPG may be an effective therapeutic strategy in DIPG and that adding a γ-secretase inhibitor during radiation therapy may be efficacious initially or during reirradiation.


Subject(s)
Brain Stem Neoplasms/pathology , Glioma/pathology , Pons/pathology , Radiation Tolerance/physiology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Blotting, Western , Brain Stem Neoplasms/metabolism , Fluorescent Antibody Technique , Glioma/metabolism , Humans , Mice , Mice, Nude , N-Myc Proto-Oncogene Protein , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Pons/metabolism , Xenograft Model Antitumor Assays
4.
Nat Med ; 21(6): 555-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25939062

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is a fatal childhood cancer. We performed a chemical screen in patient-derived DIPG cultures along with RNA-seq analyses and integrated computational modeling to identify potentially effective therapeutic strategies. The multi-histone deacetylase inhibitor panobinostat demonstrated therapeutic efficacy both in vitro and in DIPG orthotopic xenograft models. Combination testing of panobinostat and the histone demethylase inhibitor GSK-J4 revealed that the two had synergistic effects. Together, these data suggest a promising therapeutic strategy for DIPG.


Subject(s)
Benzazepines/administration & dosage , Brain Stem Neoplasms/drug therapy , Glioma/drug therapy , Hydroxamic Acids/administration & dosage , Indoles/administration & dosage , Pyrimidines/administration & dosage , Animals , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology , Disease Models, Animal , Drug Synergism , Glioma/genetics , Glioma/pathology , Humans , Panobinostat , Sequence Analysis, RNA , Xenograft Model Antitumor Assays
5.
Oncotarget ; 6(5): 3165-77, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25638158

ABSTRACT

Atypical teratoid rhabdoid tumor (AT/RT) is among the most fatal of all pediatric brain tumors. Aside from loss of function mutations in the SMARCB1 (BAF47/INI1/SNF5) chromatin remodeling gene, little is known of other molecular drivers of AT/RT. LIN28A and LIN28B are stem cell factors that regulate thousands of RNAs and are expressed in aggressive cancers. We identified high-levels of LIN28A and LIN28B in AT/RT primary tumors and cell lines, with corresponding low levels of the LIN28-regulated microRNAs of the let-7 family. Knockdown of LIN28A by lentiviral shRNA in the AT/RT cell lines CHLA-06-ATRT and BT37 inhibited growth, cell proliferation and colony formation and induced apoptosis. Suppression of LIN28A in orthotopic xenograft models led to a more than doubling of median survival compared to empty vector controls (48 vs 115 days). LIN28A knockdown led to increased expression of let-7b and let-7g microRNAs and a down-regulation of KRAS mRNA. AT/RT primary tumors expressed increased mitogen activated protein (MAP) kinase pathway activity, and the MEK inhibitor selumetinib (AZD6244) decreased AT/RT growth and increased apoptosis. These data implicate LIN28/RAS/MAP kinase as key drivers of AT/RT tumorigenesis and indicate that targeting this pathway may be a therapeutic option in this aggressive pediatric malignancy.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Protein Kinase Inhibitors/pharmacology , RNA-Binding Proteins/metabolism , Rhabdoid Tumor/drug therapy , Teratoma/drug therapy , Animals , Apoptosis , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Targeted Therapy , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , RNA Interference , RNA-Binding Proteins/genetics , Rhabdoid Tumor/enzymology , Rhabdoid Tumor/genetics , Teratoma/enzymology , Teratoma/genetics , Time Factors , Transfection , Tumor Burden , Xenograft Model Antitumor Assays
6.
J Neuropathol Exp Neurol ; 74(2): 177-85, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25575139

ABSTRACT

Atypical teratoid/rhabdoid tumor (AT/RT) is an aggressive pediatric central nervous system tumor. The poor prognosis of AT/RT warrants identification of novel therapeutic targets and strategies. High-mobility Group AT-hook 2 (HMGA2) is a developmentally important chromatin-modifying protein that positively regulates tumor growth, self-renewal, and invasion in other cancer types. High-mobility group A2 was recently identified as being upregulated in AT/RT tissue, but the role of HMGA2 in brain tumors remains unknown. We used lentiviral short-hairpin RNA to suppress HMGA2 in AT/RT cell lines and found that loss of HMGA2 led to decreased cell growth, proliferation, and colony formation and increased apoptosis. We also found that suppression of HMGA2 negatively affected in vivo orthotopic xenograft tumor growth, more than doubling median survival of mice from 58 days to 153 days. Our results indicate a role for HMGA2 in AT/RT in vitro and in vivo and demonstrate that HMGA2 is a potential therapeutic target in these lethal pediatric tumors.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , HMGA2 Protein/metabolism , Rhabdoid Tumor/pathology , Teratoma/pathology , Animals , Apoptosis/genetics , Apoptosis/physiology , Carcinogenicity Tests , Cell Line, Tumor , Cell Proliferation/genetics , Colony-Forming Units Assay , Disease Models, Animal , Gene Expression Regulation, Neoplastic/physiology , HMGA2 Protein/genetics , Humans , Kaplan-Meier Estimate , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Xenograft Model Antitumor Assays
7.
Neuro Oncol ; 17(1): 70-80, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25053853

ABSTRACT

BACKGROUND: 3-bromopyruvate (3-BrPA) and dichloroacetate (DCA) are inhibitors of cancer-cell specific aerobic glycolysis. Their application in glioma is limited by 3-BrPA's inability to cross the blood-brain-barrier and DCA's dose-limiting toxicity. The safety and efficacy of intracranial delivery of these compounds were assessed. METHODS: Cytotoxicity of 3-BrPA and DCA were analyzed in U87, 9L, and F98 glioma cell lines. 3-BrPA and DCA were incorporated into biodegradable pCPP:SA wafers, and the maximally tolerated dose was determined in F344 rats. Efficacies of the intracranial 3-BrPA wafer and DCA wafer were assessed in a rodent allograft model of high-grade glioma, both as a monotherapy and in combination with temozolomide (TMZ) and radiation therapy (XRT). RESULTS: 3-BrPA and DCA were found to have similar IC50 values across the 3 glioma cell lines. 5% 3-BrPA wafer-treated animals had significantly increased survival compared with controls (P = .0027). The median survival of rats with the 50% DCA wafer increased significantly compared with both the oral DCA group (P = .050) and the controls (P = .02). Rats implanted on day 0 with a 5% 3-BrPA wafer in combination with TMZ had significantly increased survival over either therapy alone. No statistical difference in survival was noted when the wafers were added to the combination therapy of TMZ and XRT, but the 5% 3-BrPA wafer given on day 0 in combination with TMZ and XRT resulted in long-term survivorship of 30%. CONCLUSION: Intracranial delivery of 3-BrPA and DCA polymer was safe and significantly increased survival in an animal model of glioma, a potential novel therapeutic approach. The combination of intracranial 3-BrPA and TMZ provided a synergistic effect.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/metabolism , Dichloroacetic Acid/administration & dosage , Glioblastoma/metabolism , Glycolysis/drug effects , Pyruvates/administration & dosage , Absorbable Implants , Animals , Antineoplastic Agents/toxicity , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/toxicity , Brain Neoplasms/prevention & control , Brain Neoplasms/radiotherapy , Cell Line, Tumor , Cell Proliferation/drug effects , Combined Modality Therapy , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Dacarbazine/toxicity , Dichloroacetic Acid/toxicity , Drug Administration Routes , Drug Combinations , Drug Delivery Systems , Female , Glioblastoma/prevention & control , Glioblastoma/radiotherapy , Humans , Kaplan-Meier Estimate , Polymers , Pyruvates/toxicity , Rats , Rats, Inbred F344 , Temozolomide
8.
Neuro Oncol ; 15(12): 1604-14, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24203892

ABSTRACT

BACKGROUND: Previous studies support a role for mitogen-activated protein kinase pathway signaling, and more recently Akt/mammalian target of rapamycin (mTOR), in pediatric low-grade glioma (PLGG), including pilocytic astrocytoma (PA). Here we further evaluate the role of the mTORC1/mTORC2 pathway in order to better direct pharmacologic blockade in these common childhood tumors. METHODS: We studied 177 PLGGs and PAs using immunohistochemistry and tested the effect of mTOR blockade on 2 PLGG cell lines (Res186 and Res259) in vitro. RESULTS: Moderate (2+) to strong (3+) immunostaining was observed for pS6 in 107/177 (59%) PAs and other PLGGs, while p4EBP1 was observed in 35/115 (30%), pElF4G in 66/112 (59%), mTOR (total) in 53/113 (47%), RAPTOR (mTORC1 component) in 64/102 (63%), RICTOR (mTORC2 component) in 48/101 (48%), and pAkt (S473) in 63/103 (61%). Complete phosphatase and tensin homolog protein loss was identified in only 7/101 (7%) of cases. In PA of the optic pathways, compared with other anatomic sites, there was increased immunoreactivity for pS6, pElF4G, mTOR (total), RICTOR, and pAkt (P < .05). We also observed increased pS6 (P = .01), p4EBP1 (P = .029), and RICTOR (P = .05) in neurofibromatosis type 1 compared with sporadic tumors. Treatment of the PLGG cell lines Res186 (PA derived) and Res259 (diffuse astrocytoma derived) with the rapalog MK8669 (ridaforolimus) led to decreased mTOR pathway activation and growth. CONCLUSIONS: These findings suggest that the mTOR pathway is active in PLGG but varies by clinicopathologic subtype. Additionally, our data suggest that mTORC2 is differentially active in optic pathway and neurofibromatosis type 1-associated gliomas. MTOR represents a potential therapeutic target in PLGG that merits further investigation.


Subject(s)
Astrocytoma/pathology , Glioma/pathology , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adolescent , Adult , Aged , Astrocytoma/metabolism , Blotting, Western , Cell Proliferation , Child , Child, Preschool , Female , Follow-Up Studies , Glioma/metabolism , Humans , Immunoenzyme Techniques , Infant , Male , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Middle Aged , Neoplasm Grading , Prognosis , Retrospective Studies , Tumor Cells, Cultured , Young Adult
9.
Oncotarget ; 4(7): 1050-64, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23846349

ABSTRACT

The cellular reprogramming factor LIN28A promotes tumorigenicity in cancers arising outside the central nervous system, but its role in brain tumors is unknown. We detected LIN28A protein in a subset of human gliomas observed higher expression in glioblastoma (GBM) than in lower grade tumors. Knockdown of LIN28A using lentiviral shRNA in GBM cell lines inhibited their invasion, growth and clonogenicity. Expression of LIN28A in GBM cell lines increased the number and size of orthotopic xenograft tumors. LIN28A expression also enhanced the invasiveness of GBM cells in vitro and in vivo. Increasing LIN28A was associated with down-regulation of tumor suppressing microRNAs let-7b and let-7g and up-regulation of the chromatin modifying protein HMGA2. The increase in tumor cell aggressiveness in vivo and in vitro was accompanied by an upregulation of pro-invasive gene expression, including SNAI1. To further investigate the oncogenic potential of LIN28A, we infected hNSC with lentiviruses encoding LIN28A together with dominant negative R248W-TP53, constitutively active KRAS and hTERT. Resulting subclones proliferated at an increased rate and formed invasive GBM-like tumors in orthotopic xenografts in immunodeficient mice. Similar to LIN28A-transduced GBM neurosphere lines, hNSC-derived tumor cells showed increased expression of HMGA2. Taken together, these data suggest a role for LIN28A in high grade gliomas and illustrate an HMGA2-associated, pro-invasive program that can be activated in GBM by LIN28A-mediated suppression of let-7 microRNAs.


Subject(s)
Brain Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/genetics , Glioblastoma/genetics , Neoplastic Stem Cells/pathology , Neural Stem Cells/pathology , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Carcinogenesis/genetics , Cell Transformation, Neoplastic/pathology , DNA-Binding Proteins/metabolism , Down-Regulation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioblastoma/metabolism , Glioblastoma/pathology , HEK293 Cells , Humans , Mice , Mice, Nude , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , RNA-Binding Proteins , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL