Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Neurobiol ; 56(6): 4566-4581, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30353492

ABSTRACT

The role of astrocytes is becoming increasingly important to understanding how glioblastoma (GBM) tumor cells diffusely invade the brain. Yet, little is known of the contribution of extracellular vesicle (EV) signaling in GBM/astrocyte interactions. We modeled GBM-EV signaling to normal astrocytes in vitro to assess whether this mode of intercellular communication could support GBM progression. EVs were isolated and characterized from three patient-derived GBM stem cells (NES+/CD133+) and their differentiated (diff) progeny cells (NES-/CD133-). Uptake of GBM-EVs by normal primary astrocytes was confirmed by fluorescence microscopy, and changes in astrocyte podosome formation and gelatin degradation were measured. Quantitative mass spectrometry-based proteomics was performed on GBM-EV stimulated astrocytes. Interaction networks were generated from common, differentially abundant proteins using Ingenuity® (Qiagen Bioinformatics) and predicted upstream regulators were tested by qPCR assays. Podosome formation and Cy3-gelatin degradation were induced in astrocytes following 24-h exposure to GBM-stem and -diff EVs, with EVs released by GBM-stem cells eliciting a greater effect. More than 1700 proteins were quantified, and bioinformatics predicted activations of MYC, NFE2L2, FN1, and TGFß1 and inhibition of TP53 in GBM-EV stimulated astrocytes that were then confirmed by qPCR. Further qPCR studies identified significantly decreased Δ133p53 and increased p53ß in astrocytes exposed to GBM-EVs that might indicate the acquisition of a pro-inflammatory, tumor-promoting senescence-associated secretory phenotype (SASP). Inhibition of TP53 and activation of MYC signaling pathways in normal astrocytes exposed to GBM-EVs may be a mechanism by which GBM manipulates astrocytes to acquire a phenotype that promotes tumor progression.


Subject(s)
Astrocytes/metabolism , Brain Neoplasms/metabolism , Extracellular Vesicles/metabolism , Glioblastoma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Aged , Cell Differentiation , Cell Line, Tumor , Cellular Senescence , Extracellular Vesicles/ultrastructure , Gelatin/metabolism , Humans , Male , Middle Aged , Nanoparticles/ultrastructure , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Particle Size , Phenotype , Podosomes/metabolism , Protein Isoforms/metabolism , Proteolysis , Proteome/metabolism
2.
Oncogenesis ; 6(5): e336, 2017 May 15.
Article in English | MEDLINE | ID: mdl-28504693

ABSTRACT

High-grade glioma (HGG) is an incurable brain cancer. The transcriptomes of cells within HGG tumors are highly heterogeneous. This renders the tumors unresponsive or able to adapt to therapeutics targeted at single pathways, thereby causing treatment failure. To overcome this, we focused on cyclin-dependent kinase 7 (CDK7), a ubiquitously expressed molecule involved in two major drivers of HGG pathogenesis: cell cycle progression and RNA polymerase-II-based transcription. We tested the activity of THZ1, an irreversible CDK7 inhibitor, on patient-derived primary HGG cell lines and ex vivo HGG patient tissue slices, using proliferation assays, microarray analysis, high-resolution respirometry, cell cycle analysis and in vivo tumor orthografts. The cellular processes affected by CDK7 inhibition were analyzed by reverse transcriptase-quantitative PCR, western blot, flow cytometry and immunofluorescence. THZ1 perturbed the transcriptome and disabled CDK activation, leading to cell cycle arrest at G2 and DNA damage. THZ1 halted transcription of the nuclear-encoded mitochondrial ribosomal genes, reducing mitochondrial translation and oxidative respiration. It also inhibited the expression of receptor tyrosine kinases such as epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor-α (PDGFR-α), reducing signaling flux through the AKT, extracellular-signal-regulated kinase 1/2 (ERK1/2) and signal transducer and activator of transcription 3 (STAT3) downstream pathways. Finally, THZ1 disrupted nucleolar, Cajal body and nuclear speckle formation, resulting in reduced cytosolic translation and malfunction of the spliceosome and thus leading to aberrant mRNA processing. These findings indicate that CDK7 is crucial for gliomagenesis, validate CDK7 as a therapeutic target and provide new insight into the cellular processes that are affected by THZ1 and induce antitumor activity.

3.
Leukemia ; 31(8): 1779-1787, 2017 08.
Article in English | MEDLINE | ID: mdl-27922598

ABSTRACT

The human EphA3 gene was discovered in a pre-B acute lymphoblastic leukemia (pre-B-ALL) using the EphA3-specific monoclonal antibody (mAb), IIIA4, which binds and activates both human and mouse EphA3. We use two models of human pre-B-ALL to examine EphA3 function, demonstrating effects on pre-B-cell receptor signaling. In therapeutic targeting studies, we demonstrated antitumor effects of the IIIA4 mAb in EphA3-expressing leukemic xenografts and no antitumor effect in the xenografts with no EphA3 expression providing evidence that EphA3 is a functional therapeutic target in pre-B-ALL. Here we show that the therapeutic effect of the anti-EphA3 antibody was greatly enhanced by adding an α-particle-emitting 213Bismuth payload.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptor, EphA3/immunology , Animals , Bismuth , Cell Line, Tumor , Humans , Immunotherapy , Mice , Receptor, EphA3/metabolism , Xenograft Model Antitumor Assays
4.
Br J Cancer ; 114(2): 188-98, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26657652

ABSTRACT

BACKGROUND: Resistance to temozolomide (TMZ) greatly limits chemotherapeutic effectiveness in glioblastoma (GBM). Here we analysed the ability of the Inhibitor-of-apoptosis-protein (IAP) antagonist birinapant to enhance treatment responses to TMZ in both commercially available and patient-derived GBM cells. METHODS: Responses to TMZ and birinapant were analysed in a panel of commercial and patient-derived GBM cell lines using colorimetric viability assays, flow cytometry, morphological analysis and protein expression profiling of pro- and antiapoptotic proteins. Responses in vivo were analysed in an orthotopic xenograft GBM model. RESULTS: Single-agent treatment experiments categorised GBM cells into TMZ-sensitive cells, birinapant-sensitive cells, and cells that were insensitive to either treatment. Combination treatment allowed sensitisation to therapy in only a subset of resistant GBM cells. Cell death analysis identified three principal response patterns: Type A cells that readily activated caspase-8 and cell death in response to TMZ while addition of birinapant further sensitised the cells to TMZ-induced cell death; Type B cells that readily activated caspase-8 and cell death in response to birinapant but did not show further sensitisation with TMZ; and Type C cells that showed no significant cell death or moderately enhanced cell death in the combined treatment paradigm. Furthermore, in vivo, a Type C patient-derived cell line that was TMZ-insensitive in vitro and showed a strong sensitivity to TMZ and TMZ plus birinapant treatments. CONCLUSIONS: Our results demonstrate remarkable differences in responses of patient-derived GBM cells to birinapant single and combination treatments, and suggest that therapeutic responses in vivo may be greatly affected by the tumour microenvironment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Brain Neoplasms/pathology , Dacarbazine/analogs & derivatives , Dipeptides/pharmacology , Glioblastoma/pathology , Indoles/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Animals , Blotting, Western , Caspase 8/drug effects , Caspase 8/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dacarbazine/pharmacology , Drug Resistance, Neoplasm/drug effects , Flow Cytometry , Humans , In Vitro Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Microscopy, Phase-Contrast , Neoplasm Transplantation , Temozolomide , Xenograft Model Antitumor Assays
5.
Br J Cancer ; 111(7): 1255-61, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25144626

ABSTRACT

The dismal outlook for patients with the most aggressive and common form of adult brain cancer, glioblastoma (GBM), motivates a search for new therapeutic strategies and targets for this aggressive disease. Here we review the findings to date on the role of Eph family receptor tyrosine kinases and their ephrin ligands in brain cancer. Expression of the Eph family of cell surface proteins is generally downregulated to very low levels in normal adult tissues making them particularly attractive for directed therapeutic targeting. Recent Eph targeting studies in pre-clinical models of GBM have been very encouraging and may provide an avenue to treat these highly refractory aggressive tumours.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Receptors, Eph Family/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Drug Screening Assays, Antitumor , Ephrins/physiology , Glioblastoma/drug therapy , Humans , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Signal Transduction
6.
Eur J Biochem ; 256(3): 550-9, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9780231

ABSTRACT

The plasminogen activator inhibitor type 2 (PAI-2) gene encodes a serine proteinase inhibitor (serpin) which is rapidly induced in response to the inflammatory cytokine, tumour necrosis factor-alpha (TNFalpha) in monocytes and macrophages. As an initial step towards understanding the molecular mechanisms underlying PAI-2 gene regulation in monocytes, we report here the analysis of the chromatin structure of 9.6 kb of 5' flanking region of the human PAI-2 gene for potential cis-acting regulatory regions using DNase I hypersensitivity mapping. Sites sensitive to DNase I were mapped in two monocytic cell lines representative of early monocytic differentiation; U937 cells, which synthesise low constitutive levels of PAI-2 that were induced following treatment with TNFalpha, and a MonoMac6 cell line which did not synthesise PAI-2 even after treatment with TNFalpha. Six DNase I hypersensitive sites (DHS) were identified; three upstream of the transcription initiation site (DH1, DH2, DH3) and three downstream of the transcription initiation site which were contained within intron A (DH4, DH5) and the exon 2/intron B junction (DH6). Among these, one distally located DH site (DH2) disappeared in both cell lines following treatment with TNFalpha. Two DH sites (DH1, DH6) were absent in PAI-2-producing U937 cells, but were present in MonoMac6 cells, which did not produce PAI-2, indicating the possible involvement of negative regulatory elements in the suppression of PAI-2 gene expression. The results demonstrate the involvement of chromatin structure in transcriptional responsiveness of the PAI-2 gene promoter and identify several loci which may be key control regions for PAI-2 gene transcription.


Subject(s)
Deoxyribonuclease I/metabolism , Monocytes/drug effects , Plasminogen Activator Inhibitor 2/genetics , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Base Sequence , Cell Line , DNA/metabolism , HL-60 Cells , Humans , Macrophages/drug effects , Molecular Sequence Data
7.
Gene ; 134(2): 201-8, 1993 Dec 08.
Article in English | MEDLINE | ID: mdl-8262378

ABSTRACT

In order to investigate the molecular basis for the regulated expression of plasminogen activator inhibitor type 2 (PAI-2), we sought to identify monocyte-derived nuclear factors which interact with the PAI-2 gene. We have explored the application of Southwestern blot mapping as an approach for identifying specific DNA-protein interactions and targeting potential regulatory DNA elements. The procedure involves an initial global screening of a crude preparation of nuclear proteins with radiolabelled DNA fragments (200-300 bp) derived from a large region (8.8 kb) of PAI-2. The bound DNA fragments are eluted and their location within PAI-2 mapped by Southern blot hybridization analysis. We have used this procedure to examine the differential binding of nuclear factors from the U937 monocytic cell in the absence and in the presence of the differentiating agent, 12-phorbol 13-myristate acetate (PMA), in order to identify proteins that bind specifically to the 5' flanking promoter region and first intron of PAI-2. Eleven DNA-binding proteins ranging in molecular mass from 27 to 92 kDa were identified, and the results define three regions of the gene which contain DNA-binding sites which may be involved in the transcriptional regulation of PAI-2. Deletion analysis using a series of 5' deletion mutants spanning PAI-2 fused to a chloramphenicol acetyltransferase-encoding reporter gene (cat) demonstrates that two of the regions identified by Southwestern blot mapping contain elements which can function to modulate PAI-2 expression in transient transfections of U937 cells.


Subject(s)
DNA-Binding Proteins/metabolism , Plasminogen Activator Inhibitor 2/genetics , Base Sequence , Cell Line , DNA/metabolism , Gene Expression Regulation , Humans , Immunoblotting , Introns , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Restriction Mapping , Tetradecanoylphorbol Acetate/pharmacology , Transfection
8.
Anaesth Intensive Care ; 10(2): 169, 1982 May.
Article in English | MEDLINE | ID: mdl-7103049
SELECTION OF CITATIONS
SEARCH DETAIL
...