Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cell Rep ; 41(3): 111511, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36261010

ABSTRACT

Glioblastoma (GBM) is characterized by extensive microvascular hyperproliferation. In addition to supplying blood to the tumor, GBM vessels also provide trophic support to glioma cells and serve as conduits for migration into the surrounding brain, promoting recurrence. Here, we enrich CD31-expressing glioma vascular cells (GVCs) and A2B5-expressing glioma tumor cells (GTCs) from primary GBM and use RNA sequencing to create a comprehensive molecular interaction map of the secreted and extracellular factors elaborated by GVCs that can interact with receptors and membrane molecules on GTCs. To validate our findings, we utilize functional assays, including a hydrogel-based migration assay and in vivo mouse models to demonstrate that one identified factor, the little-studied integrin binding sialoprotein (IBSP), enhances tumor growth and promotes the migration of GTCs along the vasculature. This perivascular niche interactome will serve as a resource to the research community in defining the potential functions of the GBM vasculature.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Animals , Mice , Glioblastoma/pathology , Integrin-Binding Sialoprotein/metabolism , Brain Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Glioma/pathology , Cell Movement , Hydrogels
2.
Cancer Res Commun ; 2(9): 1049-1060, 2022 09.
Article in English | MEDLINE | ID: mdl-36213002

ABSTRACT

Analysis of tumor gene expression is an important approach for the classification and identification of therapeutic vulnerabilities. However, targeting glioblastoma (GBM) based on molecular subtyping has not yet translated into successful therapies. Here, we present an integrative approach based on molecular pathways to expose new potentially actionable targets. We used gene set enrichment analysis (GSEA) to conduct an unsupervised clustering analysis to condense the gene expression data from bulk patient samples and patient-derived gliomasphere lines into new gene signatures. We identified key targets that are predicted to be differentially activated between tumors and were functionally validated in a library of gliomasphere cultures. Resultant cluster-specific gene signatures associated not only with hallmarks of cell cycle and stemness gene expression, but also with cell-type specific markers and different cellular states of GBM. Several upstream regulators, such as PIK3R1 and EBF1 were differentially enriched in cells bearing stem cell like signatures and bear further investigation. We identified the transcription factor E2F1 as a key regulator of tumor cell proliferation and self-renewal in only a subset of gliomasphere cultures predicted to be E2F1 signaling dependent. Our in vivo work also validated the functional significance of E2F1 in tumor formation capacity in the predicted samples. E2F1 inhibition also differentially sensitized E2F1-dependent gliomasphere cultures to radiation treatment. Our findings indicate that this novel approach exploring cancer pathways highlights key therapeutic vulnerabilities for targeting GBM.


Subject(s)
Glioblastoma , Humans , Glioblastoma/drug therapy , Cell Line, Tumor , Transcription Factors/metabolism , Cell Proliferation/genetics , E2F1 Transcription Factor/genetics
3.
Dev Cell ; 57(1): 32-46.e8, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35016005

ABSTRACT

We test the hypothesis that glioblastoma harbors quiescent cancer stem cells that evade anti-proliferative therapies. Functional characterization of spontaneous glioblastomas from genetically engineered mice reveals essential quiescent stem-like cells that can be directly isolated from tumors. A derived quiescent cancer-stem-cell-specific gene expression signature is enriched in pre-formed patient GBM xenograft single-cell clusters that lack proliferative gene expression. A refined human 118-gene signature is preserved in quiescent single-cell populations from primary and recurrent human glioblastomas. The F3 cell-surface receptor mRNA, expressed in the conserved signature, identifies quiescent tumor cells by antibody immunohistochemistry. F3-antibody-sorted glioblastoma cells exhibit stem cell gene expression, enhance self-renewal in culture, drive tumor initiation and serial transplantation, and reconstitute tumor heterogeneity. Upon chemotherapy, the spared cancer stem cell pool becomes activated and accelerates transition to proliferation. These results help explain conventional treatment failure and lay a conceptual framework for alternative therapies.


Subject(s)
Cell Survival/physiology , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle/genetics , Cell Division/physiology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/pathology , Heterografts , Humans , Mice , Neoplasm Invasiveness/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Transcriptome/genetics
4.
Cell Stem Cell ; 28(8): 1397-1410.e4, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34010628

ABSTRACT

NF1-associated malignant peripheral nerve sheath tumors (MPNSTs) are the major cause of mortality in neurofibromatosis. MPNSTs arise from benign peripheral nerve plexiform neurofibromas that originate in the embryonic neural crest cell lineage. Using reporter transgenes that label early neural crest lineage cells in multiple NF1 MPNST mouse models, we discover and characterize a rare MPNST cell population with stem-cell-like properties, including quiescence, that is essential for tumor initiation and relapse. Following isolation of these cells, we derive a cancer-stem-cell-specific gene expression signature that includes consensus embryonic neural crest genes and identify Nestin as a marker for the MPNST cell of origin. Combined targeting of cancer stem cells along with antimitotic chemotherapy yields effective tumor inhibition and prolongs survival. Enrichment of the cancer stem cell signature in cognate human tumors supports the generality and relevance of cancer stem cells to MPNST therapy development.


Subject(s)
Neurofibromatosis 1 , Neurofibrosarcoma , Animals , Disease Models, Animal , Mice , Neoplasm Recurrence, Local , Neurofibromatosis 1/genetics
5.
Proc Natl Acad Sci U S A ; 117(49): 31448-31458, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229571

ABSTRACT

Adult neural stem cells (NSC) serve as a reservoir for brain plasticity and origin for certain gliomas. Lineage tracing and genomic approaches have portrayed complex underlying heterogeneity within the major anatomical location for NSC, the subventricular zone (SVZ). To gain a comprehensive profile of NSC heterogeneity, we utilized a well-validated stem/progenitor-specific reporter transgene in concert with single-cell RNA sequencing to achieve unbiased analysis of SVZ cells from infancy to advanced age. The magnitude and high specificity of the resulting transcriptional datasets allow precise identification of the varied cell types embedded in the SVZ including specialized parenchymal cells (neurons, glia, microglia) and noncentral nervous system cells (endothelial, immune). Initial mining of the data delineates four quiescent NSC and three progenitor-cell subpopulations formed in a linear progression. Further evidence indicates that distinct stem and progenitor populations reside in different regions of the SVZ. As stem/progenitor populations progress from neonatal to advanced age, they acquire a deficiency in transition from quiescence to proliferation. Further data mining identifies stage-specific biological processes, transcription factor networks, and cell-surface markers for investigation of cellular identities, lineage relationships, and key regulatory pathways in adult NSC maintenance and neurogenesis.


Subject(s)
Aging/genetics , Cell Lineage , Lateral Ventricles/anatomy & histology , Lateral Ventricles/cytology , Stem Cell Niche/genetics , Transcriptome/genetics , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Biomarkers/metabolism , Cell Lineage/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Transgenes
6.
Cancer Cell ; 38(3): 366-379.e8, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32649888

ABSTRACT

Glioblastoma, the predominant adult malignant brain tumor, has been computationally classified into molecular subtypes whose functional relevance remains to be comprehensively established. Tumors from genetically engineered glioblastoma mouse models initiated by identical driver mutations in distinct cells of origin portray unique transcriptional profiles reflective of their respective lineage. Here, we identify corresponding transcriptional profiles in human glioblastoma and describe patient-derived xenografts with species-conserved subtype-discriminating functional properties. The oligodendrocyte lineage-associated glioblastoma subtype requires functional ERBB3 and harbors unique therapeutic sensitivities. These results highlight the importance of cell lineage in glioblastoma independent of driver mutations and provide a methodology for functional glioblastoma classification for future clinical investigations.


Subject(s)
Brain Neoplasms/genetics , Cell Lineage/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Dasatinib/pharmacology , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Kaplan-Meier Estimate , Mice, Knockout , Mice, Nude , Oligodendroglia/cytology , Oligodendroglia/metabolism , Xenograft Model Antitumor Assays/methods
7.
Neuro Oncol ; 20(6): 764-775, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29136244

ABSTRACT

Background: Clinical trials of therapies directed against nodes of the signaling axis of phosphatidylinositol-3 kinase/Akt/mammalian target of rapamycin (mTOR) in glioblastoma (GBM) have had disappointing results. Resistance to mTOR inhibitors limits their efficacy. Methods: To determine mechanisms of resistance to chronic mTOR inhibition, we performed tandem screens on patient-derived GBM cultures. Results: An unbiased phosphoproteomic screen quantified phosphorylation changes associated with chronic exposure to the mTOR inhibitor rapamycin, and our analysis implicated a role for glycogen synthase kinase (GSK)3B attenuation in mediating resistance that was confirmed by functional studies. A targeted short hairpin RNA screen and further functional studies both in vitro and in vivo demonstrated that microtubule-associated protein (MAP)1B, previously associated predominantly with neurons, is a downstream effector of GSK3B-mediated resistance. Furthermore, we provide evidence that chronic rapamycin induces microtubule stability in a MAP1B-dependent manner in GBM cells. Additional experiments explicate a signaling pathway wherein combinatorial extracellular signal-regulated kinase (ERK)/mTOR targeting abrogates inhibitory phosphorylation of GSK3B, leads to phosphorylation of MAP1B, and confers sensitization. Conclusions: These data portray a compensatory molecular signaling network that imparts resistance to chronic mTOR inhibition in primary, human GBM cell cultures and points toward new therapeutic strategies.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/pathology , Microtubule-Associated Proteins/metabolism , RNA, Small Interfering/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioblastoma/drug therapy , Glioblastoma/genetics , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
PLoS One ; 11(11): e0164649, 2016.
Article in English | MEDLINE | ID: mdl-27855170

ABSTRACT

We present here a novel genetic algorithm-based random forest (GARF) modeling technique that enables a reduction in the complexity of large gene disease signatures to highly accurate, greatly simplified gene panels. When applied to 803 glioblastoma multiforme samples, this method allowed the 840-gene Verhaak et al. gene panel (the standard in the field) to be reduced to a 48-gene classifier, while retaining 90.91% classification accuracy, and outperforming the best available alternative methods. Additionally, using this approach we produced a 32-gene panel which allows for better consistency between RNA-seq and microarray-based classifications, improving cross-platform classification retention from 69.67% to 86.07%. A webpage producing these classifications is available at http://simplegbm.semel.ucla.edu.


Subject(s)
Brain Neoplasms/genetics , Computational Biology/methods , Gene Expression Profiling , Glioblastoma/genetics , Transcriptome , Algorithms , Brain Neoplasms/mortality , Datasets as Topic , Gene Expression Profiling/methods , Genomics/methods , Glioblastoma/mortality , Humans , Kaplan-Meier Estimate , Molecular Sequence Annotation , Prognosis , Reproducibility of Results , Web Browser
9.
Mol Cancer Ther ; 15(6): 1271-8, 2016 06.
Article in English | MEDLINE | ID: mdl-27196770

ABSTRACT

Inhibition of both the de novo (DNP) and salvage (NSP) pathways of nucleoside synthesis has been demonstrated to impair leukemia cells. We endeavored to determine whether this approach would be efficacious in glioblastoma. To diminish nucleoside biosynthesis, we utilized compound DI-39, which selectively targets NSP, in combination with thymidine (dT), which selectively targets DNP. We employed in vitro and ex vivo models to determine the effects of pretreatment with dT + DI-39 on brain tumor stem cells (BTSC). Here, we demonstrate that this combinatorial therapy elicits a differential response across a spectrum of human patient-derived glioblastoma cultures. As determined by apoptotic markers, most cultures were relatively resistant to treatment, although a subset was highly sensitive. Sensitivity was unrelated to S-phase delay and to DNA damage induced by treatment. Bioinformatics analysis indicated that response across cultures was associated with the transcription factor PAX3 (associated with resistance) and with canonical pathways, including the nucleotide excision repair pathway, PTEN (associated with resistance), PI3K/AKT (associated with sensitivity), and ErbB2-ErbB3. Our in vitro assays demonstrated that, in sensitive cultures, clonal sphere formation was reduced upon removal from pretreatment. In contrast, in a resistant culture, clonal sphere formation was slightly increased upon removal from pretreatment. Moreover, in an intracranial xenograft model, pretreatment of a sensitive culture caused significantly smaller and fewer tumors. In a resistant culture, tumors were equivalent irrespective of pretreatment. These results indicate that, in the subset of sensitive glioblastoma, BTSCs are targeted by inhibition of pyrimidine synthesis. Mol Cancer Ther; 15(6); 1271-8. ©2016 AACR.


Subject(s)
Brain Neoplasms/drug therapy , Deoxycytidine Kinase/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Glioblastoma/drug therapy , Neoplastic Stem Cells/drug effects , Pyrimidines/administration & dosage , Sulfonamides/administration & dosage , Thymidine/administration & dosage , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Glioblastoma/metabolism , Humans , Mice , PAX3 Transcription Factor/metabolism , PTEN Phosphohydrolase/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , Thymidine/pharmacology , Xenograft Model Antitumor Assays
10.
Neuro Oncol ; 18(10): 1367-78, 2016 10.
Article in English | MEDLINE | ID: mdl-27116978

ABSTRACT

BACKGROUND: Gliomasphere cultures are widely utilized for the study of glioblastoma (GBM). However, this model system is not well characterized, and the utility of current classification methods is not clear. METHODS: We used 71 gliomasphere cultures from 68 individuals. Using gene expression-based classification, we performed unsupervised clustering and associated gene expression with gliomasphere phenotypes and patient survival. RESULTS: Some aspects of the gene expression-based classification method were robust because the gliomasphere cultures retained their classification over many passages, and IDH1 mutant gliomaspheres were all proneural. While gene expression of a subset of gliomasphere cultures was more like the parent tumor than any other tumor, gliomaspheres did not always harbor the same classification as their parent tumor. Classification was not associated with whether a sphere culture was derived from primary or recurrent GBM or associated with the presence of EGFR amplification or rearrangement. Unsupervised clustering of gliomasphere gene expression distinguished 2 general categories (mesenchymal and nonmesenchymal), while multidimensional scaling distinguished 3 main groups and a fourth minor group. Unbiased approaches revealed that PI3Kinase, protein kinase A, mTOR, ERK, Integrin, and beta-catenin pathways were associated with in vitro measures of proliferation and sphere formation. Associating gene expression with gliomasphere phenotypes and patient outcome, we identified genes not previously associated with GBM: PTGR1, which suppresses proliferation, and EFEMP2 and LGALS8, which promote cell proliferation. CONCLUSIONS: This comprehensive assessment reveals advantages and limitations of using gliomaspheres to model GBM biology, and provides a novel strategy for selecting genes for future study.


Subject(s)
Gene Expression Profiling/methods , Glioblastoma/genetics , Tumor Cells, Cultured , Blotting, Western , Cell Culture Techniques/methods , Cluster Analysis , Gene Regulatory Networks , Glioblastoma/classification , Glioblastoma/pathology , Humans , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Transcriptome
11.
Integr Mol Med ; 2(3)2015.
Article in English | MEDLINE | ID: mdl-33889422

ABSTRACT

BACKGROUND: The goals of the study were (1) to determine the impact of inorganic mercury exposure on glucose homeostasis; and (2) to evaluate the effectiveness of two community-based interventions in promoting dietary changes among American Indian college students to reduce risk factors for Type-2 Diabetes including fasting glucose, insulin, and mercury levels, weight, and body mass index. METHODS: To accomplish goal one, the National Health and Nutrition Examination Survey (NHANES) dataset was analyzed using a previously published method to determine if there is a relationship between inorganic blood mercury and fasting glucose. To accomplish goal two, ten college students were recruited and randomly assigned to a group receiving the online macroepigenetics nutrition course and the support group for eliminating corn sweeteners. Participants in both groups were assessed for diet patterns, weight, body mass index (BMI), fasting glucose, insulin, and mercury levels. The interventions were implemented over a 10-week period. RESULTS: Analysis of the NHANES data (n=16,232) determined a direct relationship between inorganic mercury in blood and fasting glucose levels (p<0.001). The participants who took the online macroepigenetics nutrition intervention course significantly improved their diets (p<0.01), and fasting blood glucose levels (p<0.01) while having lower levels of inorganic mercury in their blood compared to the subjects in the group who eliminated corn sweeteners from their diet and participated in the support group. The trend in lower blood inorganic mercury was strong with p=0.052. The participants in the support group who eliminated corn sweeteners from their diet achieved significant weight loss (p<0.01) and reduced their body mass index (p<0.01). CONCLUSION: Total blood mercury levels may be influenced by dietary intake of highly processed foods and lower inorganic mercury levels are associated with lower fasting glucose levels. Alternative community-based interventions emphasizing the role food ingredients and toxic substances play in gene modulation and the development of diseases can result in significant dietary improvements and reductions in risk factors associated with type-2 diabetes. A healthier diet can be promoted among community members using a novel online nutrition course. Consumption of corn sweeteners may be a risk factor in the development of obesity.

12.
Stem Cell Reports ; 3(5): 725-34, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25418720

ABSTRACT

A period of mild brain overgrowth with an unknown etiology has been identified as one of the most common phenotypes in autism. Here, we test the hypothesis that maternal inflammation during critical periods of embryonic development can cause brain overgrowth and autism-associated behaviors as a result of altered neural stem cell function. Pregnant mice treated with low-dose lipopolysaccharide at embryonic day 9 had offspring with brain overgrowth, with a more pronounced effect in PTEN heterozygotes. Exposure to maternal inflammation also enhanced NADPH oxidase (NOX)-PI3K pathway signaling, stimulated the hyperproliferation of neural stem and progenitor cells, increased forebrain microglia, and produced abnormal autism-associated behaviors in affected pups. Our evidence supports the idea that a prenatal neuroinflammatory dysregulation in neural stem cell redox signaling can act in concert with underlying genetic susceptibilities to affect cellular responses to environmentally altered cellular levels of reactive oxygen species.


Subject(s)
Autistic Disorder/immunology , Brain/immunology , Inflammation/immunology , Prenatal Exposure Delayed Effects/immunology , Stem Cells/immunology , Animals , Animals, Newborn , Blotting, Western , Brain/metabolism , Brain/pathology , Cell Proliferation , Cells, Cultured , Female , Grooming , Inflammation/chemically induced , Inflammation/physiopathology , Lipopolysaccharides/immunology , Lipopolysaccharides/toxicity , Male , Maze Learning , Mice , Microglia/immunology , NADPH Oxidases/immunology , NADPH Oxidases/metabolism , Oxidation-Reduction , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Signal Transduction/immunology
13.
Mol Cancer Res ; 12(5): 694-702, 2014 May.
Article in English | MEDLINE | ID: mdl-24505127

ABSTRACT

UNLABELLED: Targeting amino acid metabolism has therapeutic implications for aggressive brain tumors. Asparagine is an amino acid that is synthesized by normal cells. However, some cancer cells lack asparagine synthetase (ASNS), the key enzyme for asparagine synthesis. Asparaginase (ASNase) contributes to eradication of acute leukemia by decreasing asparagine levels in serum and cerebrospinal fluid. However, leukemic cells may become ASNase-resistant by upregulating ASNS. High expression of ASNS has also been associated with biologic aggressiveness of other cancers, including gliomas. Here, the impact of enzymatic depletion of asparagine on proliferation of brain tumor cells was determined. ASNase was used as monotherapy or in combination with conventional chemotherapeutic agents. Viability assays for ASNase-treated cells demonstrated significant growth reduction in multiple cell lines. This effect was reversed by glutamine in a dose-dependent manner--as expected, because glutamine is the main amino group donor for asparagine synthesis. ASNase treatment also reduced sphere formation by medulloblastoma and primary glioblastoma cells. ASNase-resistant glioblastoma cells exhibited elevated levels of ASNS mRNA. ASNase cotreatment significantly enhanced gemcitabine or etoposide cytotoxicity against glioblastoma cells. Xenograft tumors in vivo showed no significant response to ASNase monotherapy and little response to temozolomide alone. However, combinatorial therapy with ASNase and temozolomide resulted in significant growth suppression for an extended duration of time. Taken together, these findings indicate that amino acid depletion warrants further investigation as adjunctive therapy for brain tumors. IMPLICATIONS: Findings have potential impact for providing adjuvant means to enhance brain tumor chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Asparaginase/pharmacology , Asparagine/deficiency , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Animals , Asparaginase/administration & dosage , Asparaginase/metabolism , Asparagine/metabolism , Aspartate-Ammonia Ligase/metabolism , Brain Neoplasms/enzymology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , DNA Damage , Dacarbazine/administration & dosage , Dacarbazine/pharmacology , Drug Synergism , Glioblastoma/drug therapy , Glioblastoma/enzymology , Glioblastoma/metabolism , Glioblastoma/pathology , Glutamine/pharmacology , Humans , Male , Medulloblastoma/drug therapy , Medulloblastoma/enzymology , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice , Mice, Nude , Temozolomide , Xenograft Model Antitumor Assays
15.
Neuro Oncol ; 13(6): 622-34, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21558073

ABSTRACT

Glioblastoma multiforme (GBM) is a devastating disease, and the current therapies have only palliative effect. Evidence is mounting to indicate that brain tumor stem cells (BTSCs) are a minority of tumor cells that are responsible for cancer initiation, propagation, and maintenance. Therapies that fail to eradicate BTSCs may ultimately lead to regrowth of residual BTSCs. However, BTSCs are relatively resistant to the current treatments. Development of novel therapeutic strategies that effectively eradicate BTSC are, therefore, essential. In a previous study, we used patient-derived GBM sphere cells (stemlike GBM cells) to enrich for BTSC and identified maternal embryonic leucine-zipper kinase (MELK) as a key regulator of survival of stemlike GBM cells in vitro. Here, we demonstrate that a thiazole antibiotic, siomycin A, potently reduced MELK expression and inhibited tumor growth in vivo. Treatment of stemlike GBM cells with siomycin A resulted in arrested self-renewal, decreased invasion, and induced apoptosis but had little effect on growth of the nonstem cells of matched tumors or normal neural stem/progenitor cells. MELK overexpression partially rescued the phenotype of siomycin A-treated stemlike GBM cells. In vivo, siomycin A pretreatment abraded the sizes of stemlike GBM cell-derived tumors in immunodeficient mice. Treatment with siomycin A of mice harboring intracranial tumors significantly prolonged their survival period compared with the control mice. Together, this study may be the first model to partially target stemlike GBM cells through a MELK-mediated pathway with siomycin A to pave the way for effective treatment of GBM.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/drug therapy , Glioblastoma/genetics , Neoplastic Stem Cells/drug effects , Peptides/therapeutic use , Protein Serine-Threonine Kinases/genetics , Animals , Apoptosis/drug effects , Blotting, Western , Brain/metabolism , Brain/pathology , Brain Neoplasms/pathology , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Flow Cytometry , Glioblastoma/pathology , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Stem Cells/metabolism , Survival Rate , Tumor Cells, Cultured
16.
J Transplant ; 2011: 740673, 2011.
Article in English | MEDLINE | ID: mdl-21559259

ABSTRACT

Background. Dose-dependent response makes certain pediatric brain tumors appropriate targets for high-dose chemotherapy with autologous hematopoietic stem-cell rescue (HDCT-AHSCR). Methods. The clinical outcomes and toxicities were analyzed retrospectively for 18 consecutive patients ≤19 y/o treated with HDCT-AHSCR at UCLA (1999-2009). Results. Patients' median age was 2.3 years. Fourteen had primary and 4 recurrent tumors: 12 neural/embryonal (7 medulloblastomas, 4 primitive neuroectodermal tumors, and a pineoblastoma), 3 glial/mixed, and 3 germ cell tumors. Eight patients had initial gross-total and seven subtotal resections. HDCT mostly consisted of carboplatin and/or thiotepa ± etoposide (n = 16). Nine patients underwent a single AHSCR and nine ≥3 tandems. Three-year progression-free and overall survival probabilities were 60.5% ± 16 and 69.3% ± 11.5. Ten patients with pre-AHSCR complete remissions were alive/disease-free, whereas 5 of 8 with measurable disease were deceased (median followup: 2.3 yrs). Nine of 13 survivors avoided radiation. Single AHSCR regimens had greater toxicity than ≥3 AHSCR (P < .01). Conclusion. HDCT-AHSCR has a definitive, though limited role for selected pediatric brain tumors with poor prognosis and pretransplant complete/partial remissions.

17.
Cancer Res ; 70(15): 6128-38, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20631065

ABSTRACT

The clinical practice of oncology is being transformed by molecular diagnostics that will enable predictive and personalized medicine. Current technologies for quantitation of the cancer proteome are either qualitative (e.g., immunohistochemistry) or require large sample sizes (e.g., flow cytometry). Here, we report a microfluidic platform-microfluidic image cytometry (MIC)-capable of quantitative, single-cell proteomic analysis of multiple signaling molecules using only 1,000 to 2,800 cells. Using cultured cell lines, we show simultaneous measurement of four critical signaling proteins (EGFR, PTEN, phospho-Akt, and phospho-S6) within the oncogenic phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway. To show the clinical application of the MIC platform to solid tumors, we analyzed a panel of 19 human brain tumor biopsies, including glioblastomas. Our MIC measurements were validated by clinical immunohistochemistry and confirmed the striking intertumoral and intratumoral heterogeneity characteristic of glioblastoma. To interpret the multiparameter, single-cell MIC measurements, we adapted bioinformatic methods including self-organizing maps that stratify patients into clusters that predict tumor progression and patient survival. Together with bioinformatic analysis, the MIC platform represents a robust, enabling in vitro molecular diagnostic technology for systems pathology analysis and personalized medicine.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Microfluidic Analytical Techniques/methods , Brain Neoplasms/metabolism , Cell Line, Tumor , ErbB Receptors/metabolism , Glioblastoma/metabolism , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/metabolism , Microfluidic Analytical Techniques/instrumentation , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reproducibility of Results , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases
18.
Pediatr Blood Cancer ; 55(4): 644-51, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20589659

ABSTRACT

BACKGROUND: Cultured brain tumors can form neurospheres harboring tumorigenic cells with self renewal and differentiation capacities. Renewable neurosphere formation has clinical predictive value in adult malignant gliomas, yet its prognostic role for pediatric brain tumors is unknown. METHODS: Established neurosphere conditions were used for culturing samples from glial, embryonal and mixed glioneuronal tumors from 56 pediatric patients. Potential associations between neurosphere formation and clinical outcome were analyzed retrospectively. RESULTS: Thirty-seven percent of all samples formed renewable neurospheres. Analysis of available clinical outcome data from 51 patients demonstrated significantly increased hazard ratios (HR) for both disease progression (HR = 9.9, P < 0.001) and death (HR = 16.6, P < 0.01) in the neurosphere forming group. Furthermore, neurosphere formation correlated with adverse progression free survival (PFS) in glial and embryonal tumors, but not in mixed glioneuronal tumors. Overall survival (OS) was significantly worse for neurosphere-forming patients with embryonal tumors, as a group and amongst the subgroup with medulloblastoma, but not in the glial group. Multivariate analysis showed that neurosphere formation was associated with diminished PFS and OS independent of age, gender, or treatment. Neurosphere formation was an independent predictor of diminished PFS of glial tumors after adjusting for grade. Multivariate analysis, adjusting for both Ki67 staining and neurosphere formation, demonstrated that neurosphere formation remained predictive of progression whereas Ki67 did not. CONCLUSIONS: Neurosphere formation is more predictive of pediatric brain tumor progression than semi-quantitative Ki67 staining. Pediatric brain tumor derived neurospheres may provide a predictive model for preclinical explorations.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Neoplasms, Germ Cell and Embryonal/pathology , Adolescent , Adult , Brain Neoplasms/mortality , Cell Line, Tumor , Child , Child, Preschool , Female , Glioma/mortality , Humans , Infant , Male , Neoplasms, Germ Cell and Embryonal/mortality , Prognosis , Proportional Hazards Models
19.
Med Hypotheses ; 74(4): 698-701, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19914008

ABSTRACT

Previous studies have demonstrated that the pituitary is a main target for inorganic mercury (I-Hg) deposition and accumulation within the brain. My recent study of the US population (1999-2006) has uncovered a significant, inverse relationship between chronic mercury exposure and levels of luteinizing hormone (LH). This association with LH signifies more than its presumed role as bioindicator for pituitary neurosecretion and function. LH is the only hormone with a rare and well characterized, high affinity binding site for mercury. On its catalytic beta subunit, LH has the structure to preferentially bind inorganic mercury almost irreversibly, and, by that manner, accumulate the neurotoxic element. Thus, it is likely that LH is an early and significant target of chronic mercury exposure. Moreover, due to the role of LH in immune-modulation and neurogenesis, I present LH as a central candidate to elucidate a causal mechanism for chronic mercury exposure and associated disease.


Subject(s)
Luteinizing Hormone/chemistry , Luteinizing Hormone/metabolism , Mercury/chemistry , Pituitary Gland/chemistry , Pituitary Gland/metabolism , Binding Sites , Female , Humans
20.
Biometals ; 22(6): 1103-14, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19697139

ABSTRACT

The purpose of this study was to assess chronic mercury exposure within the US population. Time trends were analyzed for blood inorganic mercury (I-Hg) levels in 6,174 women, ages 18-49, in the NHANES, 1999-2006 data sets. Multivariate logistic regression distinguished a significant, direct correlation within the US population between I-Hg detection and years since the start of the survey (OR = 1.49, P < 0.001). Within this population, I-Hg detection rose sharply from 2% in 1999-2000 to 30% in 2005-2006. In addition, the population averaged mean I-Hg concentration rose significantly over that same period from 0.33 to 0.39 µ/L (Anova, P < 0.001). In a separate analysis, multivariate logistic regression indicated that I-Hg detection was significantly associated with age (OR = 1.02, P < 0.001). Furthermore, multivariate logistic regression revealed significant associations of both I-Hg detection and mean concentration with biomarkers for the main targets of mercury deposition and effect: the liver, immune system, and pituitary. This study provides compelling evidence that I-Hg deposition within the human body is a cumulative process, increasing with age and in the population over time, since 1999, as a result of chronic mercury exposure. Furthermore, our results indicate that I-Hg deposition is associated with the significant biological markers for main targets of exposure, deposition, and effect. Accumulation of focal I-Hg deposits within the human body due to chronic mercury exposure provides a mechanism which suggests a time dependent rise in the population risks for associated disease.


Subject(s)
Mercury Compounds/toxicity , Mercury/toxicity , Nutrition Surveys/trends , Adolescent , Adult , Age Factors , Bilirubin/analysis , Biomarkers/analysis , Female , Humans , Immune System/cytology , Leukocyte Count , Liver/chemistry , Logistic Models , Luteinizing Hormone/analysis , Mercury/analysis , Mercury/pharmacokinetics , Mercury Compounds/analysis , Mercury Compounds/pharmacokinetics , Middle Aged , Pituitary Gland/chemistry , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...