Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Neuro Oncol ; 25(8): 1415-1427, 2023 08 03.
Article in English | MEDLINE | ID: mdl-36705543

ABSTRACT

BACKGROUND: The lack of murine glioblastoma models that mimic the immunobiology of human disease has impeded basic and translational immunology research. We, therefore, developed murine glioblastoma stem cell lines derived from Nestin-CreERT2QkL/L; Trp53L/L; PtenL/L (QPP) mice driven by clinically relevant genetic mutations common in human glioblastoma. This study aims to determine the immune sensitivities of these QPP lines in immunocompetent hosts and their underlying mechanisms. METHODS: The differential responsiveness of QPP lines was assessed in the brain and flank in untreated, anti-PD-1, or anti-CTLA-4 treated mice. The impact of genomic landscape on the responsiveness of each tumor was measured through whole exome sequencing. The immune microenvironments of sensitive (QPP7) versus resistant (QPP8) lines were compared in the brain using flow cytometry. Drivers of flank sensitivity versus brain resistance were also measured for QPP8. RESULTS: QPP lines are syngeneic to C57BL/6J mice and demonstrate varied sensitivities to T cell immune checkpoint blockade ranging from curative responses to complete resistance. Infiltrating tumor immune analysis of QPP8 reveals improved T cell fitness and augmented effector-to-suppressor ratios when implanted subcutaneously (sensitive), which are absent on implantation in the brain (resistant). Upregulation of PD-L1 across the myeloid stroma acts to establish this state of immune privilege in the brain. In contrast, QPP7 responds to checkpoint immunotherapy even in the brain likely resulting from its elevated neoantigen burden. CONCLUSIONS: These syngeneic QPP models of glioblastoma demonstrate clinically relevant profiles of immunotherapeutic sensitivity and potential utility for both mechanistic discovery and evaluation of immune therapies.


Subject(s)
Glioblastoma , Humans , Animals , Mice , Glioblastoma/pathology , Mice, Inbred C57BL , Immunotherapy/methods , T-Lymphocytes/metabolism , Tumor Microenvironment
2.
Front Cell Dev Biol ; 10: 931387, 2022.
Article in English | MEDLINE | ID: mdl-36051438

ABSTRACT

Glioblastoma (GBM) is a high-grade, aggressive brain tumor with dismal median survival time of 15 months. Chromosome 6q (Ch6q) is a hotspot of genomic alterations, which is commonly deleted or hyper-methylated in GBM. Two neighboring genes in this region, QKI and PRKN have been appointed as tumor suppressors in GBM. While a genetically modified mouse model (GEMM) of GBM has been successfully generated with Qk deletion in the central nervous system (CNS), in vivo genetic evidence supporting the tumor suppressor function of Prkn has not been established. In the present study, we generated a mouse model with Prkn-null allele and conditional Trp53 and Pten deletions in the neural stem cells (NSCs) and compared the tumorigenicity of this model to our previous GBM model with Qk deletion within the same system. We find that Qk but not Prkn is the potent tumor suppressor in the frequently altered Ch6q region in GBM.

3.
JCI Insight ; 7(12)2022 06 22.
Article in English | MEDLINE | ID: mdl-35653194

ABSTRACT

Novel therapeutic strategies targeting glioblastoma (GBM) often fail in the clinic, partly because preclinical models in which hypotheses are being tested do not recapitulate human disease. To address this challenge, we took advantage of our previously developed spontaneous Qk/Trp53/Pten (QPP) triple-knockout model of human GBM, comparing the immune microenvironment of QPP mice with that of patient-derived tumors to determine whether this model provides opportunity for gaining insights into tumor physiopathology and preclinical evaluation of therapeutic agents. Immune profiling analyses and single-cell sequencing of implanted and spontaneous tumors from QPP mice and from patients with glioma revealed intratumoral immune components that were predominantly myeloid cells (e.g., monocytes, macrophages, and microglia), with minor populations of T, B, and NK cells. When comparing spontaneous and implanted mouse samples, we found more neutrophils and T and NK cells in the implanted model. Neutrophils and T and NK cells were increased in abundance in samples derived from human high-grade glioma compared with those derived from low-grade glioma. Overall, our data demonstrate that our implanted and spontaneous QPP models recapitulate the immunosuppressive myeloid-dominant nature of the tumor microenvironment of human gliomas. Our model provides a suitable tool for investigating the complex immune compartment of gliomas.


Subject(s)
Glioblastoma , Glioma , Animals , Disease Models, Animal , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Macrophages , Mice , Tumor Microenvironment
4.
Elife ; 102021 05 04.
Article in English | MEDLINE | ID: mdl-33942715

ABSTRACT

Myelination depends on timely, precise control of oligodendrocyte differentiation and myelinogenesis. Cholesterol is the most abundant component of myelin and essential for myelin membrane assembly in the central nervous system. However, the underlying mechanisms of precise control of cholesterol biosynthesis in oligodendrocytes remain elusive. In the present study, we found that Qki depletion in neural stem cells or oligodendrocyte precursor cells in neonatal mice resulted in impaired cholesterol biosynthesis and defective myelinogenesis without compromising their differentiation into Aspa+Gstpi+ myelinating oligodendrocytes. Mechanistically, Qki-5 functions as a co-activator of Srebp2 to control transcription of the genes involved in cholesterol biosynthesis in oligodendrocytes. Consequently, Qki depletion led to substantially reduced concentration of cholesterol in mouse brain, impairing proper myelin assembly. Our study demonstrated that Qki-Srebp2-controlled cholesterol biosynthesis is indispensable for myelinogenesis and highlights a novel function of Qki as a transcriptional co-activator beyond its canonical function as an RNA-binding protein.


Subject(s)
Biosynthetic Pathways/genetics , Cholesterol/biosynthesis , Cholesterol/genetics , Gene Expression Regulation , Myelin Sheath/physiology , RNA-Binding Proteins/genetics , Sterol Regulatory Element Binding Protein 2/genetics , Animals , Animals, Newborn , Cell Differentiation , Mice , Neurogenesis , Transcription Factors
5.
Nat Commun ; 12(1): 3005, 2021 05 21.
Article in English | MEDLINE | ID: mdl-34021134

ABSTRACT

Defective cholesterol biosynthesis in eye lens cells is often associated with cataracts; however, how genes involved in cholesterol biosynthesis are regulated in lens cells remains unclear. Here, we show that Quaking (Qki) is required for the transcriptional activation of genes involved in cholesterol biosynthesis in the eye lens. At the transcriptome level, lens-specific Qki-deficient mice present downregulation of genes associated with the cholesterol biosynthesis pathway, resulting in a significant reduction of total cholesterol level in the eye lens. Mice with Qki depletion in lens epithelium display progressive accumulation of protein aggregates, eventually leading to cataracts. Notably, these defects are attenuated by topical sterol administration. Mechanistically, we demonstrate that Qki enhances cholesterol biosynthesis by recruiting Srebp2 and Pol II in the promoter regions of cholesterol biosynthesis genes. Supporting its function as a transcription co-activator, we show that Qki directly interacts with single-stranded DNA. In conclusion, we propose that Qki-Srebp2-mediated cholesterol biosynthesis is essential for maintaining the cholesterol level that protects lens from cataract development.


Subject(s)
Cholesterol/biosynthesis , Lens, Crystalline/metabolism , RNA-Binding Proteins/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Animals , Cell Line , Gene Products, pol , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones , RNA, Messenger , RNA-Binding Proteins/genetics , Sterol Regulatory Element Binding Protein 2/genetics
6.
Gastroenterology ; 161(1): 196-210, 2021 07.
Article in English | MEDLINE | ID: mdl-33745946

ABSTRACT

BACKGROUND & AIMS: Understanding the mechanisms by which tumors adapt to therapy is critical for developing effective combination therapeutic approaches to improve clinical outcomes for patients with cancer. METHODS: To identify promising and clinically actionable targets for managing colorectal cancer (CRC), we conducted a patient-centered functional genomics platform that includes approximately 200 genes and paired this with a high-throughput drug screen that includes 262 compounds in four patient-derived xenografts (PDXs) from patients with CRC. RESULTS: Both screening methods identified exportin 1 (XPO1) inhibitors as drivers of DNA damage-induced lethality in CRC. Molecular characterization of the cellular response to XPO1 inhibition uncovered an adaptive mechanism that limited the duration of response in TP53-mutated, but not in TP53-wild-type CRC models. Comprehensive proteomic and transcriptomic characterization revealed that the ATM/ATR-CHK1/2 axes were selectively engaged in TP53-mutant CRC cells upon XPO1 inhibitor treatment and that this response was required for adapting to therapy and escaping cell death. Administration of KPT-8602, an XPO1 inhibitor, followed by AZD-6738, an ATR inhibitor, resulted in dramatic antitumor effects and prolonged survival in TP53-mutant models of CRC. CONCLUSIONS: Our findings anticipate tremendous therapeutic benefit and support the further evaluation of XPO1 inhibitors, especially in combination with DNA damage checkpoint inhibitors, to elicit an enduring clinical response in patients with CRC harboring TP53 mutations.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Biomarkers, Tumor/genetics , Colorectal Neoplasms/drug therapy , Karyopherins/antagonists & inhibitors , Mutation , Protein Kinase Inhibitors/administration & dosage , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Databases, Genetic , HCT116 Cells , HT29 Cells , Humans , Indoles/administration & dosage , Karyopherins/metabolism , Mice , Morpholines/administration & dosage , Piperazines/administration & dosage , Pyridines/administration & dosage , Pyrimidines/administration & dosage , Receptors, Cytoplasmic and Nuclear/metabolism , Sulfonamides/administration & dosage , Xenograft Model Antitumor Assays , Exportin 1 Protein
7.
J Exp Med ; 218(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-33045062

ABSTRACT

The mechanism underpinning the regulation of microglial phagocytosis in demyelinating diseases is unclear. Here, we showed that the Quaking protein (Qki) in microglia was greatly induced by demyelination in the brains of both mice and humans. Deletion of the Quaking gene (Qk) in microglia severely impaired the clearance of myelin debris. Transcriptomic profiling indicated that depletion of Qki impaired total RNA levels and splicing of the genes involved in phagosome formation and maturation. RNA immunoprecipitation (RIP) confirmed the physical interactions between the Qki protein and the mRNAs of Qki targets that are involved in phagocytosis, indicating that Qki regulates their RNA stability. Both Qki depletion and inhibition of Qki target Cd36 greatly reduced the phagocytic activity of microglia and macrophages. The defective uptake and degradation of myelin debris caused by Qki depletion in microglia resulted in unresolved myelin debris that impaired axon integrity, oligodendrocyte maturation, and subsequent remyelination. Thus, our results demonstrate that Qki is an essential regulator of microglia's phagocytic activity under demyelinating conditions.


Subject(s)
Demyelinating Autoimmune Diseases, CNS/metabolism , Microglia/metabolism , Phagocytosis , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Animals , Axons/metabolism , Axons/pathology , CD36 Antigens/genetics , CD36 Antigens/metabolism , Demyelinating Autoimmune Diseases, CNS/genetics , Demyelinating Autoimmune Diseases, CNS/pathology , Humans , Mice , Mice, Transgenic , Microglia/pathology , Myelin Sheath/genetics , Myelin Sheath/metabolism , Myelin Sheath/pathology , Phagosomes/genetics , Phagosomes/metabolism , Phagosomes/pathology , RNA, Messenger/genetics , RNA-Binding Proteins/genetics
8.
J Clin Invest ; 130(5): 2220-2236, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32202512

ABSTRACT

Lipid-rich myelin forms electrically insulating, axon-wrapping multilayers that are essential for neural function, and mature myelin is traditionally considered metabolically inert. Surprisingly, we discovered that mature myelin lipids undergo rapid turnover, and quaking (Qki) is a major regulator of myelin lipid homeostasis. Oligodendrocyte-specific Qki depletion, without affecting oligodendrocyte survival, resulted in rapid demyelination, within 1 week, and gradually neurological deficits in adult mice. Myelin lipids, especially the monounsaturated fatty acids and very-long-chain fatty acids, were dramatically reduced by Qki depletion, whereas the major myelin proteins remained intact, and the demyelinating phenotypes of Qki-depleted mice were alleviated by a high-fat diet. Mechanistically, Qki serves as a coactivator of the PPARß-RXRα complex, which controls the transcription of lipid-metabolism genes, particularly those involved in fatty acid desaturation and elongation. Treatment of Qki-depleted mice with PPARß/RXR agonists significantly alleviated neurological disability and extended survival durations. Furthermore, a subset of lesions from patients with primary progressive multiple sclerosis were characterized by preferential reductions in myelin lipid contents, activities of various lipid metabolism pathways, and expression level of QKI-5 in human oligodendrocytes. Together, our results demonstrate that continuous lipid synthesis is indispensable for mature myelin maintenance and highlight an underappreciated role of lipid metabolism in demyelinating diseases.


Subject(s)
DNA-Binding Proteins/metabolism , Demyelinating Diseases/metabolism , Lipid Metabolism , Myelin Sheath/metabolism , PPAR-beta/metabolism , RNA-Binding Proteins/metabolism , Animals , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Fatty Acids/genetics , Fatty Acids/metabolism , Humans , Mice , Mice, Knockout , Myelin Sheath/genetics , Myelin Sheath/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , PPAR-beta/antagonists & inhibitors , PPAR-beta/genetics , RNA-Binding Proteins/genetics
9.
Nat Genet ; 49(1): 75-86, 2017 01.
Article in English | MEDLINE | ID: mdl-27841882

ABSTRACT

Stem cells, including cancer stem cells (CSCs), require niches to maintain stemness, yet it is unclear how CSCs maintain stemness in the suboptimal environment outside their niches during invasion. Postnatal co-deletion of Pten and Trp53 in mouse neural stem cells (NSCs) leads to the expansion of these cells in their subventricular zone (SVZ) niches but fails to maintain stemness outside the SVZ. We discovered that Qki is a major regulator of NSC stemness. Qk deletion on a Pten-/-; Trp53-/- background helps NSCs maintain their stemness outside the SVZ in Nes-CreERT2; QkL/L; PtenL/L; Trp53L/L mice, which develop glioblastoma with a penetrance of 92% and a median survival time of 105 d. Mechanistically, Qk deletion decreases endolysosome-mediated degradation and enriches receptors essential for maintaining self-renewal on the cytoplasmic membrane to cope with low ligand levels outside niches. Thus, downregulation of endolysosome levels by Qki loss helps glioma stem cells (GSCs) maintain their stemness in suboptimal environments outside their niches.


Subject(s)
Brain Neoplasms/pathology , Endosomes/metabolism , Glioma/pathology , Lysosomes/metabolism , Neoplastic Stem Cells/pathology , Neural Stem Cells/pathology , RNA-Binding Proteins/physiology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cells, Cultured , Female , Glioma/genetics , Glioma/metabolism , Mice , Mice, Knockout , Mice, Nude , Mice, SCID , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , PTEN Phosphohydrolase/physiology , Proteolysis , Receptors, Cell Surface/metabolism , Stem Cell Niche , Tumor Suppressor Protein p53/physiology
10.
Stem Cells Int ; 2016: 7849890, 2016.
Article in English | MEDLINE | ID: mdl-26880988

ABSTRACT

Glioblastoma remains the most common and devastating primary brain tumor despite maximal therapy with surgery, chemotherapy, and radiation. The glioma stem cell (GSC) subpopulation has been identified in glioblastoma and likely plays a key role in resistance of these tumors to conventional therapies as well as recurrent disease. GSCs are capable of self-renewal and differentiation; glioblastoma-derived GSCs are capable of de novo tumor formation when implanted in xenograft models. Further, GSCs possess unique surface markers, modulate characteristic signaling pathways to promote tumorigenesis, and play key roles in glioma vascular formation. These features, in addition to microenvironmental factors, present possible targets for specifically directing therapy against the GSC population within glioblastoma. In this review, the authors summarize the current knowledge of GSC biology and function and the role of GSCs in new vascular formation within glioblastoma and discuss potential therapeutic approaches to target GSCs.

11.
J Transl Med ; 14: 46, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26861698

ABSTRACT

BACKGROUND: The majority of glioblastomas have aberrant receptor tyrosine kinase (RTK)/RAS/phosphoinositide 3 kinase (PI3K) signaling pathways and malignant glioma cells are thought to be addicted to these signaling pathways for their survival and proliferation. However, recent studies suggest that monotherapies or inappropriate combination therapies using the molecular targeted drugs have limited efficacy possibly because of tumor heterogeneities, signaling redundancy and crosstalk in intracellular signaling network, indicating necessity of rationale and methods for efficient personalized combination treatments. Here, we evaluated the growth of colonies obtained from glioma tumor-initiating cells (GICs) derived from glioma sphere culture (GSC) in agarose and examined the effects of combination treatments on GICs using targeted drugs that affect the signaling pathways to which most glioma cells are addicted. METHODS: Human GICs were cultured in agarose and treated with inhibitors of RTKs, non-receptor kinases or transcription factors. The colony number and volume were analyzed using a colony counter, and Chou-Talalay combination indices were evaluated. Autophagy and apoptosis were also analyzed. Phosphorylation of proteins was evaluated by reverse phase protein array and immunoblotting. RESULTS: Increases of colony number and volume in agarose correlated with the Gompertz function. GICs showed diverse drug sensitivity, but inhibitions of RTK and RAF/MEK or PI3K by combinations such as EGFR inhibitor and MEK inhibitor, sorafenib and U0126, erlotinib and BKM120, and EGFR inhibitor and sorafenib showed synergy in different subtypes of GICs. Combination of erlotinib and sorafenib, synergistic in GSC11, induced apoptosis and autophagic cell death associated with suppressed Akt and ERK signaling pathways and decreased nuclear PKM2 and ß-catenin in vitro, and tended to improve survival of nude mice bearing GSC11 brain tumor. Reverse phase protein array analysis of the synergistic treatment indicated involvement of not only MEK and PI3K signaling pathways but also others associated with glucose metabolism, fatty acid metabolism, gene transcription, histone methylation, iron transport, stress response, cell cycle, and apoptosis. CONCLUSION: Inhibiting RTK and RAF/MEK or PI3K could induce synergistic cytotoxicity but personalization is necessary. Examining colonies in agarose initiated by GICs from each patient may be useful for drug sensitivity testing in personalized cancer therapy.


Subject(s)
Glioma/drug therapy , Glioma/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neoplastic Stem Cells/pathology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/therapeutic use , raf Kinases/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Autophagy/drug effects , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Humans , Inhibitory Concentration 50 , Male , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , raf Kinases/metabolism
12.
J Vis Exp ; (98)2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25938254

ABSTRACT

Telomere dysfunction-induced loss of genome integrity and its associated DNA damage signaling and checkpoint responses are well-established drivers that cause tissue degeneration during ageing. Cancer, with incidence rates greatly increasing with age, is characterized by short telomere lengths and high telomerase activity. To study the roles of telomere dysfunction and telomerase reactivation in ageing and cancer, the protocol shows how to generate two murine inducible telomerase knock-in alleles 4-Hydroxytamoxifen (4-OHT)-inducible TERT-Estrogen Receptor (mTERT-ER) and Lox-Stopper-LoxTERT (LSL-mTERT). The protocol describes the procedures to induce telomere dysfunction and reactivate telomerase activity in mTERT-ER and LSL-mTERT mice in vivo. The representative data show that reactivation of telomerase activity can ameliorate the tissue degenerative phenotypes induced by telomere dysfunction. In order to determine the impact of telomerase reactivation on tumorigenesis, we generated prostate tumor model G4 PB-Cre4 Pten(L/L) p53(L/L) LSL-mTERT(L/L) and thymic T-cell lymphoma model G4 Atm(-/-) mTERT(ER/ER). The representative data show that telomerase reactivation in the backdrop of genomic instability induced by telomere dysfunction can greatly enhance tumorigenesis. The protocol also describes the procedures used to isolate neural stem cells (NSCs) from mTERT-ER and LSL-mTERT mice and reactivate telomerase activity in NSCs in vitro. The representative data show that reactivation of telomerase can enhance the self-renewal capability and neurogenesis in vitro. Finally, the protocol describes the procedures for performing telomere FISH (Fluorescence In Situ Hybridization) on both mouse FFPE (Formalin Fixed and Paraffin Embedded) brain tissues and metaphase chromosomes of cultured cells.


Subject(s)
Alleles , Neoplasms/genetics , Regeneration/genetics , Telomerase/genetics , Animals , Cells, Cultured , Female , Gene Knock-In Techniques/methods , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred C57BL , Neoplasms/pathology , Neural Stem Cells/cytology , Neural Stem Cells/enzymology , Telomere/metabolism
13.
J Biol Chem ; 286(37): 32843-53, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21795717

ABSTRACT

Recent studies suggest that a small subpopulation of malignant cells with stem-like properties is resistant to chemotherapy and may be responsible for the existence of residual cancer after treatment. We have isolated highly tumorigenic cancer cells with 100-fold increase in tumor initiating capacity from the tumor xenografts of human glioblastoma U87 cells in mice. These cells exhibit stem-like properties and show unique energy metabolic characteristics including low mitochondrial respiration, increased glycolysis for ATP generation, and preference for hypoxia to maintain their stemness and tumor forming capacity. Mechanistically, mitochondrial depression in the highly tumorigenic cells occurs mainly at complex II of the electron transport chain with a down-regulation of the succinate dehydrogenase subunit B, leading to deregulation of hypoxia-inducible factors. Under hypoxia, the stem-like cancer cells are resistant to conventional anticancer agents but are sensitive to glycolytic inhibition. Furthermore, combination of glycolytic inhibition with standard therapeutic agents is effective in killing the tumor-initiating cells in vitro and inhibits tumor formation in vivo. Our study suggests that stem-like cancer cells prefer a low oxygen microenvironment and actively utilize the glycolytic pathway for ATP generation. Inhibition of glycolysis may be an effective strategy to eradicate residual cancer stem cells that are otherwise resistant to chemotherapeutic agents in their hypoxic niches.


Subject(s)
Adenosine Triphosphate/biosynthesis , Glioblastoma/metabolism , Glycolysis , Animals , Cell Hypoxia , Cell Line, Tumor , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Mice , Mice, Nude , Mitochondria/enzymology , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Neoplasm, Residual , Oxygen/metabolism , Succinate Dehydrogenase/metabolism , Transcription Factors/metabolism , Transplantation, Heterologous
14.
Int J Cancer ; 128(4): 787-96, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-20473884

ABSTRACT

Polynuclear platinum compounds are more effective at killing glioblastoma cells than cisplatin, work by a different mechanism, and typically do not induce high levels of apoptosis at early time points after exposure. Here, we tested the hypothesis that combining BBR3610, the most potent polynuclear platinum, with a phosphoinositide-3-kinase (PI3K) inhibitor would promote apoptosis and enhance the impact on glioblastoma cells. The PI3K pathway is commonly activated in glioblastoma and promotes tumor cell survival, suggesting that its inhibition would make cells more sensitive to cytotoxic agents. We chose PX-866 as a PI3K inhibitor as it is a clinically promising agent being evaluated for brain tumor therapy. Combining BBR3610 and PX-866 resulted in synergistic killing of cultured glioma cells and an extension of survival in an orthotopic xenograft animal model. Both agents alone induced autophagy, and this appeared to be saturated, because when they were combined no additional autophagy was observed. However, the combination of PX-866 and BBR3610 did induce statistically significant increases in the level of apoptosis, associated with a reduction in pAkt and pBad, as well as inhibition of transwell migration. We conclude that combining polynuclear platinums with PI3K inhibitors has translational potential and alters the cellular response to include early apoptosis.


Subject(s)
Brain Neoplasms/drug therapy , Glioma/drug therapy , Gonanes/therapeutic use , Organoplatinum Compounds/therapeutic use , Animals , Apoptosis/drug effects , Autophagy/drug effects , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Drug Therapy, Combination , Glioma/metabolism , Glioma/pathology , Humans , Male , Mice , Mice, Nude , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Survival Rate , Tumor Cells, Cultured
15.
Neuro Oncol ; 12(12): 1269-77, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20713409

ABSTRACT

BBR3610 is a polynuclear platinum compound, in which two platinums are linked by a spermine-like linker, and studies in a variety of cancers, including glioma, have shown that it is more potent than conventional platinums and works by different means. Identifying the mechanism of action of BBR3610 would help in developing the drug further for clinical use. Previous work showed that BBR3610 does not induce immediate apoptosis but results in an early G2/M arrest. Here, we report that BBR3610 induces early autophagy in glioma cells. Increased autophagy was also seen in intracranial xenografts treated with BBR3610. Interestingly, upon attenuation of autophagy by RNAi-mediated knockdown of ATG5 or ATG6/BECN1, no change in cell viability was observed, suggesting that the autophagy is neither an effective protection against BBR3610 nor an important part of the mechanism by which BBR3610 reduces glioma cell viability. This prompted a multimodal analysis of 4 cell lines over 2 weeks posttreatment with BBR3610, which showed that the G2/M arrest occurred early and apoptosis occurred later in all cell lines. The cells that survived entered a senescent state associated with mitotic catastrophe in 2 of the cell lines. Together, our data show that the response to treatment with a single agent is complex and changes over time.


Subject(s)
Apoptosis/drug effects , Autophagy , Brain Neoplasms/pathology , Cell Division/drug effects , G2 Phase/drug effects , Glioma/pathology , Organoplatinum Compounds/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Beclin-1 , Blotting, Western , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Cisplatin/therapeutic use , Drug Therapy, Combination , Glioma/drug therapy , Glioma/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Nude , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Autophagy ; 5(4): 537-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19270491

ABSTRACT

Anticancer therapies can induce autophagy in tumor cells and the role of autophagy in these cells may depend on the type of tumor, the stage of tumorigenesis, and the nature and extent of the insult. Appropriate modification of autophagy, that is, suppression of cell-protective autophagy or enhancement of cell-killing autophagy could augment cytotoxicity caused by anticancer therapy. Imatinib mesylate is an inhibitor of tyrosine kinases and is used for the therapy of patients with tumors including leukemias, but is not effective as a monotherapy for malignant glioma. To seek a strategy to augment the therapeutic efficacy of imatinib, we examined the mode of cytotoxicity of imatinib in human malignant glioma cells. Since imatinib induced cytotoxicity associated with autophagy, we tested the effect of inhibition of autophagy on imatinib-induced cytotoxicity. We found that imatinib-induced cytotoxicity is attenuated by inhibition of autophagy at an early stage but augmented by inhibition of autophagy at a late stage through increasing apoptosis following mitochondrial damage. Though the mechanism of the stage-specific effect of inhibiting autophagy on cytotoxicity remains to be elucidated, our findings could be useful for developing a new strategy to enhance the efficacy of anticancer therapy by modulating autophagy.


Subject(s)
Autophagy/drug effects , Neoplasm Staging , Piperazines/pharmacology , Pyrimidines/pharmacology , Benzamides , Humans , Imatinib Mesylate , Models, Biological
17.
Int J Cancer ; 124(5): 1060-71, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19048625

ABSTRACT

Malignant gliomas are common primary tumors of the central nervous system. The prognosis of patients with malignant glioma is poor in spite of current intensive therapy and thus novel therapeutic modalities are necessary. Imatinib mesylate, a tyrosine kinase inhibitor, is effective in the therapy of tumors including leukemias but not as a monotherapy for malignant glioma. Recently, it is thought that the adequate modulation of autophagy can enhance efficacy of anticancer therapy. The outcome of autophagy manipulation, however, seems to depend on the autophagy initiator, the combined stimuli, the extent of cellular damage and the type of cells, and it is not yet fully understood how we should modulate autophagy to augment efficacy of each anticancer therapy. In this study, we examined the effect of imatinib with or without different types of autophagy inhibitors on human malignant glioma cells. Imatinib inhibited the viability of U87-MG and U373-MG cells in a dose dependent manner and caused nonapoptotic autophagic cell death. Suppression of imatinib-induced autophagy by 3-methyladenine or small interfering RNA against Atg5, which inhibit autophagy at an early stage, attenuated the imatinib-induced cytotoxicity. In contrast, inhibition of autophagy at a late stage by bafilomycin A1 or RTA 203 enhanced imatinib-induced cytotoxicity through the induction of apoptosis following mitochondrial disruption. Our findings suggest that therapeutic efficiency of imatinib for malignant glioma may be augmented by inhibition of autophagy at a late stage, and that appropriate modulation of autophagy may sensitize tumor cells to anticancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Brain Neoplasms/drug therapy , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Glioma/drug therapy , Macrolides/pharmacology , Piperazines/pharmacology , Pyrimidines/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Autophagy-Related Protein 5 , Benzamides , Brain Neoplasms/pathology , Cell Line, Tumor , Cisplatin/pharmacology , Extracellular Signal-Regulated MAP Kinases/physiology , Glioma/pathology , Humans , Imatinib Mesylate , MAP Kinase Signaling System/drug effects , Membrane Potential, Mitochondrial/drug effects , Microtubule-Associated Proteins/antagonists & inhibitors , Paclitaxel/pharmacology
18.
Autophagy ; 4(4): 467-75, 2008 May.
Article in English | MEDLINE | ID: mdl-18259115

ABSTRACT

Autophagy, an evolutionarily conserved response to stress, has recently been implicated in cancer initiation and progression, but the detailed mechanisms and functions have not yet been fully elucidated. One major obstacle to our understanding is lack of an efficient and robust method to specifically monitor autophagic cells in cancer specimens. To identify molecular events associated with autophagy, we performed cDNA microarray analysis of autophagic glioblastoma cell lines. Based on the analysis, we raised a polyclonal antibody against isoform B of human microtubule-associated protein 1 light chain 3 (LC3B). Application of the anti-LC3B antibody revealed the presence of autophagic cells in both in vitro and in vivo settings. Of the 65 glioblastoma tissues, 31 had highly positive cytoplasmic staining of LC3B. The statistical interaction between cytoplasmic staining of LC3B and Karnofsky Performance Scale score was significant. High expression of LC3B was associated with an improved outcome for patients with poorer performance, whereas, for patients with normal performance, survival was better for patients with low staining than with high staining of LC3B. Anti-LC3B antibody provides a useful tool for monitoring the induction of autophagy in cancer cells and tissues.


Subject(s)
Antibodies/immunology , Antibodies/metabolism , Autophagy/physiology , Glioblastoma/pathology , Microtubule-Associated Proteins/immunology , Microtubule-Associated Proteins/metabolism , Animals , Antineoplastic Agents/metabolism , Arsenic Trioxide , Arsenicals/metabolism , Cell Line, Tumor , Ceramides/metabolism , Dacarbazine/analogs & derivatives , Dacarbazine/metabolism , Female , Glioblastoma/metabolism , Humans , Mice , Mice, Nude , Microtubule-Associated Proteins/genetics , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Oxides/metabolism , Paclitaxel/metabolism , Phagosomes/metabolism , Phagosomes/ultrastructure , Survival Rate , Temozolomide , Transplantation, Heterologous , Tubulin Modulators/metabolism
19.
Neurol Med Chir (Tokyo) ; 47(5): 222-8, 2007 May.
Article in English | MEDLINE | ID: mdl-17527050

ABSTRACT

A 51-year-old woman had been followed up for 10 years for recurrence of pilocytic astrocytoma 5 years after the initial treatment consisting of subtotal resection, chemotherapy, and radiation therapy. The patient presented with sudden onset of headache and vomiting. Computed tomography and T(2)*-weighted magnetic resonance imaging revealed hemorrhage in the tumor located in the right basal ganglia, thalamus, and hypothalamus. She underwent gross total resection of the lesion. Histological examination confirmed recurrent pilocytic astrocytoma with organizing hematoma and granulation tissue. Although neither symptomatic hemorrhage nor late benign recurrence is common, careful long-term follow up is necessary for patients with pilocytic astrocytoma.


Subject(s)
Astrocytoma/pathology , Brain Neoplasms/pathology , Cerebral Hemorrhage/etiology , Neoplasm Recurrence, Local/pathology , Adult , Astrocytoma/complications , Astrocytoma/surgery , Brain Neoplasms/complications , Brain Neoplasms/surgery , Female , Granulation Tissue/pathology , Humans , Neoplasm Recurrence, Local/complications , Neoplasm Recurrence, Local/surgery
20.
Neurol Med Chir (Tokyo) ; 45(10): 530-5, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16247240

ABSTRACT

An 88-year-old woman presented with a supratentorial primitive neuroectodermal tumor (PNET) manifesting as disturbance of consciousness and left hemiplegia. Magnetic resonance imaging showed a large mass lesion in the right frontotemporal region. She underwent biopsy of the lesion that confirmed the diagnosis of PNET. Her poor condition only allowed chemotherapy with methyl 6-[3-(2-chloroethyl)-3-nitrosoureido]-6-deoxy-alpha-D-glucopyranoside (MCNU), vincristine, and prednisolone to be performed. The patient died approximately 6 months after diagnosis due to enlargement of the tumor. Supratentorial PNET is a rare tumor, especially in adults. Multimodal therapy consisting of gross total or subtotal resection, radiation therapy, and chemotherapy is generally considered necessary for patients with supratentorial PNET. However, the condition of each patient should be considered in determining the therapeutic plan, especially in the case of extremely aged patients, since supratentorial PNET is malignant and long-term survival is rare despite aggressive treatment.


Subject(s)
Neuroectodermal Tumors, Primitive/pathology , Supratentorial Neoplasms/pathology , Aged, 80 and over , Female , Humans , Neuroectodermal Tumors, Primitive/complications , Neuroectodermal Tumors, Primitive/therapy , Supratentorial Neoplasms/complications , Supratentorial Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...