Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Acta Neuropathol Commun ; 7(1): 55, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30971321

ABSTRACT

Melanoma patients carry a high risk of developing brain metastases, and improvements in survival are still measured in weeks or months. Durable disease control within the brain is impeded by poor drug penetration across the blood-brain barrier, as well as intrinsic and acquired drug resistance. Augmented mitochondrial respiration is a key resistance mechanism in BRAF-mutant melanomas but, as we show in this study, this dependence on mitochondrial respiration may also be exploited therapeutically. We first used high-throughput pharmacogenomic profiling to identify potentially repurposable compounds against BRAF-mutant melanoma brain metastases. One of the compounds identified was ß-sitosterol, a well-tolerated and brain-penetrable phytosterol. Here we show that ß-sitosterol attenuates melanoma cell growth in vitro and also inhibits brain metastasis formation in vivo. Functional analyses indicated that the therapeutic potential of ß-sitosterol was linked to mitochondrial interference. Mechanistically, ß-sitosterol effectively reduced mitochondrial respiratory capacity, mediated by an inhibition of mitochondrial complex I. The net result of this action was increased oxidative stress that led to apoptosis. This effect was only seen in tumor cells, and not in normal cells. Large-scale analyses of human melanoma brain metastases indicated a significant role of mitochondrial complex I compared to brain metastases from other cancers. Finally, we observed completely abrogated BRAF inhibitor resistance when vemurafenib was combined with either ß-sitosterol or a functional knockdown of mitochondrial complex I. In conclusion, based on its favorable tolerability, excellent brain bioavailability, and capacity to inhibit mitochondrial respiration, ß-sitosterol represents a promising adjuvant to BRAF inhibitor therapy in patients with, or at risk for, melanoma brain metastases.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Melanoma/genetics , Melanoma/metabolism , Mitochondria/drug effects , Proto-Oncogene Proteins B-raf/genetics , Sitosterols/administration & dosage , Animals , Apoptosis/drug effects , Brain Neoplasms/complications , Cell Line, Tumor , Drug Repositioning , Female , Humans , Melanoma/complications , Mice, Transgenic , Mitochondria/metabolism , Mutation , Oxidative Stress/drug effects , Transcriptome
2.
Int J Cancer ; 144(7): 1735-1745, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30289977

ABSTRACT

Glioblastoma multiforme (GBM) has a poor prognosis with an overall survival of 14-15 months after surgery, radiation and chemotherapy using temozolomide (TMZ). A major problem is that the tumors acquire resistance to therapy. In an effort to improve the therapeutic efficacy of TMZ, we performed a genome-wide RNA interference (RNAi) synthetic lethality screen to establish a functional gene signature for TMZ sensitivity in human GBM cells. We then queried the Connectivity Map database to search for drugs that would induce corresponding changes in gene expression. By this approach we identified several potential pharmacological sensitizers to TMZ, where the most potent drug was the established antipsychotic agent Thioridazine, which significantly improved TMZ sensitivity while not demonstrating any significant toxicity alone. Mechanistically, we show that the specific chemosensitizing effect of Thioridazine is mediated by impairing autophagy, thereby preventing adaptive metabolic alterations associated with TMZ resistance. Moreover, we demonstrate that Thioridazine inhibits late-stage autophagy by impairing fusion between autophagosomes and lysosomes. Finally, Thioridazine in combination with TMZ significantly inhibits brain tumor growth in vivo, demonstrating the potential clinical benefits of compounds targeting the autophagy-lysosome pathway. Our study emphasizes the feasibility of exploiting drug repurposing for the design of novel therapeutic strategies for GBM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Autophagy/drug effects , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Temozolomide/administration & dosage , Thioridazine/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagosomes/drug effects , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Glioblastoma/genetics , Humans , Lysosomes/drug effects , Mice , Synthetic Lethal Mutations , Temozolomide/therapeutic use , Thioridazine/pharmacology , Xenograft Model Antitumor Assays
3.
Autophagy ; 14(11): 2007-2022, 2018.
Article in English | MEDLINE | ID: mdl-30025493

ABSTRACT

Flavokawain B (FKB), a natural kava chalcone, displays potent antitumor activity in various types of cancer. The mechanism of action, however, remains unclear. Here, we evaluated the efficacy of FKB in the treatment of human glioblastoma multiforme (GBM) as well as the molecular basis for its inhibitory effects in cancer. Approximately 60% of GBM cells became senescent after treatment with FKB as assessed in the senescence-associated (SA)-GLB1/SA-ß-galactosidase assay. The cellular process of autophagy potentially contributed to the establishment of senescence. Transmission electron microscopy revealed the formation of autophagic vesicles under FKB treatment, and MAP1LC3B (microtubule associated protein 1 light chain 3 beta)-II was increased. Transfection of ATG5 or ATG7 small interfering RNAs (siRNAs) inhibited FKB-induced autophagy in U251 cells. Western blot revealed that molecular components of the endoplasmic reticulum stress pathway were activated, including ATF4 (activating transcription factor 4) and DDIT3 (DNA damage inducible transcript 3), while levels of TRIB3 (tribbles pseudokinase 3) increased. In addition, based on the phosphorylation status, the AKT-MTOR-RPS6KB1 pathway was inhibited, which induced autophagy in GBM cells. Inhibition of autophagy by autophagy inhibitors 3-methyladenine and chloroquine or knockdown of ATG5 or ATG7 caused FKB-treated U251 cells to switch from senescence to apoptosis. Finally, knockdown of ATG5 or treatment with chloroquine in combination with FKB, significantly inhibited tumor growth in vivo. Our results demonstrated that FKB induced protective autophagy through the ATF4-DDIT3-TRIB3-AKT-MTOR-RPS6KB1 signaling pathway in GBM cells, indicating that the combination treatment of FKB with autophagy inhibitors may potentially be an effective therapeutic strategy for GBM. ABBREVIATIONS: 3-MA: 3-methyladenine; 4-PBA: 4-phenylbutyrate; AKT: AKT serine/threonine kinase; ATF4: activating transcription factor 4; ATG: autophagy related; CASP3: caspase 3; CCK-8: cell counting kit-8; CDKN1A: cyclin-dependent kinase inhibitor 1A; CQ: chloroquine; DDIT3: DNA damage inducible transcript 3; DMEM: Dulbecco's modified Eagle's medium; EIF2A: eukaryotic translation initiation factor 2A; EIF2AK3: eukaryotic translation initiation factor 2 alpha kinase 3; ER: endoplasmic reticulum; FKB: flavokawain B; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GBM: glioblastoma multiforme; GFP: green fluorescent protein; HSPA5: heat shock protein family A (Hsp70) member 5; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; MTOR: mechanistic target of rapamycin kinase; PARP1: poly(ADP-ribose) polymerase; 1RPS6KB1: ribosomal protein S6 kinase B1; SA-GLB1: senescence-associated galactosidase beta 1; siRNA: short interfering RNA; SQSTM1: sequestosome 1; TEM: transmission electron microscopy; TRIB3: tribbles pseudokinase 3; TUNEL: deoxynucleotidyl transferase-mediated dUTP nick-end labeling.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Autophagy/drug effects , Cell Proliferation/drug effects , Endoplasmic Reticulum Stress/drug effects , Flavonoids/pharmacology , Glioma/pathology , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Autophagy/genetics , Autophagy-Related Protein 5/antagonists & inhibitors , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 7/antagonists & inhibitors , Autophagy-Related Protein 7/genetics , Cell Proliferation/genetics , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/physiology , Flavonoids/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Glioma/drug therapy , Glioma/genetics , Glioma/metabolism , Humans , Male , Mice , Mice, Nude , Xenograft Model Antitumor Assays
4.
J Exp Clin Cancer Res ; 36(1): 118, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28870216

ABSTRACT

BACKGROUND: Resistance to adjuvant radiotherapy is a major cause of treatment failure in patients with glioblastoma (GBM). Autophagy inhibitors have been shown to enhance the efficacy of radiotherapy for certain solid tumors. However, current inhibitors do not penetrate the blood-brain-barrier (BBB). Here, we assessed the radiosensitivity effects of the antipsychotic drug trifluoperazine (TFP) on GBM in vitro and in vivo. METHODS: U251 and U87 GBM cell lines as well as GBM cells from a primary human biopsy (P3), were used in vitro and in vivo to evaluate the efficacy of TFP treatment. Viability and cytotoxicity was evaluated by CCK-8 and clonogenic formation assays. Molecular studies using immunohistochemistry, western blots, immunofluorescence and qPCR were used to gain mechanistic insight into the biological activity of TFP. Preclinical therapeutic efficacy was evaluated in orthotopic xenograft mouse models. RESULTS: IC50 values of U251, U87 and P3 cells treated with TFP were 16, 15 and 15.5 µM, respectively. TFP increased the expression of LC3B-II and p62, indicating a potential disruption of autophagy flux. These results were further substantiated by a decreased Lysotracker Red uptake, indicating impaired acidification of the lysosomes. We show that TFP and radiation had an additive effect when combined. This effect was in part due to impaired TFP-induced homologous recombination. Mechanistically we show that down-regulation of cathepsin L might explain the radiosensitivity effect of TFP. Finally, combining TFP and radiation resulted in a significant antitumor effect in orthotopic GBM xenograft models. CONCLUSIONS: This study provides a strong rationale for further clinical studies exploring the combination therapy of TFP and radiation to treat GBM patients.


Subject(s)
Autophagy/drug effects , Glioblastoma/drug therapy , Radiation Tolerance/drug effects , Trifluoperazine/administration & dosage , Animals , Autophagy/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/pathology , Glioblastoma/radiotherapy , Homologous Recombination/drug effects , Humans , Mice , Microtubule-Associated Proteins/genetics , RNA-Binding Proteins/genetics , Radiation Tolerance/genetics , Xenograft Model Antitumor Assays
5.
Oncotarget ; 7(41): 66944-66958, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27557493

ABSTRACT

There is an urgent need for new therapeutic strategies for patients with glioblastoma multiforme (GBM). Previous studies have shown that berberine (BBR), a natural plant alkaloid, has potent anti-tumor activity. However, the mechanisms leading to cancer cell death have not been clearly elucidated. In this study, we show that BBR has profound effects on the metabolic state of GBM cells, leading to high autophagy flux and impaired glycolytic capacity. Functionally, these alterations reduce the invasive properties, proliferative potential and induce apoptotic cell death. The molecular alterations preceding these changes are characterized by inhibition of the AMPK/mTOR/ULK1 pathway. Finally, we demonstrate that BBR significantly reduces tumor growth in vivo, demonstrating the potential clinical benefits for autophagy modulating plant alkaloids in cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Autophagy/drug effects , Berberine/pharmacology , Glioblastoma/pathology , Signal Transduction/drug effects , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy-Related Protein-1 Homolog/drug effects , Autophagy-Related Protein-1 Homolog/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Glioblastoma/metabolism , Humans , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Nude , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
6.
Genes Cancer ; 5(5-6): 186-200, 2014 May.
Article in English | MEDLINE | ID: mdl-25061502

ABSTRACT

Originally known as host defence peptides for their substantial bacteriotoxic effects, many cationic antimicrobial peptides also exhibit a potent cytotoxic activity against cancer cells. Their mode of action is characterized mostly by electrostatic interactions with the plasma membrane, leading to membrane disruption and rapid necrotic cell death. In this work, we have designed a novel cationic peptide of 27 amino acids (Cypep-1), which shows efficacy against a number of cancer cell types, both in vitro and in vivo, while normal human fibroblasts were significantly less affected. Surface plasmon resonance experiments as well as liposome leakage assays monitored by fluorescence spectroscopy revealed a substantial binding affinity of Cypep-1 to negatively charged liposomes and induced significant leakage of liposome content after exposure to the peptide. The observed membranolytic effect of Cypep-1 was confirmed by scanning electron microscopy (SEM) as well as by time-lapse confocal microscopy. Pharmacokinetic profiling of Cypep-1 in rats showed a short plasma half-life after i.v. injection, followed mainly by retention in the liver, spleen and kidneys. Extremely low concentrations within the organs of the central nervous system indicated that Cypep-1 did not pass the blood-brain-barrier. Local treatment of 4T1 murine mammary carcinoma allografts by means of a single local bolus injection of Cypep-1 led to a significant reduction of tumour growth in the following weeks and prolonged survival. Detailed histological analysis of the treated tumours revealed large areas of necrosis. In sum, our findings show that the novel cationic peptide Cypep-1 displays a strong cytolytic activity against cancer cells both in vitro and in vivo and thus holds a substantial therapeutic potential.

7.
Acta Neuropathol ; 125(5): 683-98, 2013 May.
Article in English | MEDLINE | ID: mdl-23429996

ABSTRACT

Angiogenesis is regarded as a hallmark of cancer progression and it has been postulated that solid tumor growth depends on angiogenesis. At present, however, it is clear that tumor cell invasion can occur without angiogenesis, a phenomenon that is particularly evident by the infiltrative growth of malignant brain tumors, such as glioblastomas (GBMs). In these tumors, amplification or overexpression of wild-type (wt) or truncated and constitutively activated epidermal growth factor receptor (EGFR) are regarded as important events in GBM development, where the complex downstream signaling events have been implicated in tumor cell invasion, angiogenesis and proliferation. Here, we show that amplification and in particular activation of wild-type EGFR represents an underlying mechanism for non-angiogenic, invasive tumor growth. Using a clinically relevant human GBM xenograft model, we show that tumor cells with EGFR gene amplification and activation diffusely infiltrate normal brain tissue independent of angiogenesis and that transient inhibition of EGFR activity by cetuximab inhibits the invasive tumor growth. Moreover, stable, long-term expression of a dominant-negative EGFR leads to a mesenchymal to epithelial-like transition and induction of angiogenic tumor growth. Analysis of human GBM biopsies confirmed that EGFR activation correlated with invasive/non-angiogenic tumor growth. In conclusion, our results indicate that activation of wild-type EGFR promotes invasion and glioblastoma development independent of angiogenesis, whereas loss of its activity results in angiogenic tumor growth.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Genes, erbB-1/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Transcriptional Activation , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Cell Culture Techniques , Cell Line, Tumor , Cetuximab , ErbB Receptors/drug effects , ErbB Receptors/genetics , Gene Amplification , Humans , Neoplasm Invasiveness/genetics , Neovascularization, Pathologic , Xenograft Model Antitumor Assays
8.
Neuro Oncol ; 15(3): 269-78, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23258843

ABSTRACT

INTRODUCTION: Glioblastoma multiforme (GBM; World Health Organization astrocytoma grade IV) is the most frequent and most malignant primary brain tumor in adults. Despite multimodal therapy, all such tumors practically recur during the course of therapy, causing a median survival of only 14.6 months in patients with newly diagnosed GBM. The present study was aimed at examining the expression of the DNA repair protein AlkB homolog 2 (ALKBH2) in human GBM and determining whether it could promote resistance to temozolomide chemotherapy. METHODS: ALKBH2 expression in GBM cell lines and in human GBM was determined by quantitative real-time PCR (qRT-PCR) and gene expression analysis, respectively. Drug sensitivity was assessed in GBM cells overexpressing ALKBH2 and in cells in which ALKBH2 expression was silenced by small-interfering (si)RNA. ALKBH2 expression following activation of the p53 pathway was examined by western blotting and qRT-PCR. RESULTS: ALKBH2 was abundantly expressed in established GBM cell lines and human GBM, and temozolomide exposure increased cellular ALKBH2 expression levels. Overexpression of ALKBH2 in the U87 and U251 GBM cell lines enhanced resistance to the methylating agents temozolomide and methyl methanesulfonate but not to the nonmethylating agent doxorubicin. Conversely, siRNA-mediated knockdown of ALKBH2 increased sensitivity of GBM cells to temozolomide and methyl methanesulfonate but not to doxorubicin or cisplatin. Nongenotoxic activation of the p53 pathway by the selective murine double minute 2 antagonist nutlin-3 caused a significant decrease in cellular ALKBH2 transcription levels. CONCLUSION: Our findings identify ALKBH2 as a novel mediator of temozolomide resistance in human GBM cells. Furthermore, we place ALKBH2 into a new cellular context by showing its regulation by the p53 pathway.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , DNA Repair Enzymes/metabolism , Dacarbazine/analogs & derivatives , Dioxygenases/metabolism , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , AlkB Homolog 2, Alpha-Ketoglutarate-Dependent Dioxygenase , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/genetics , Dacarbazine/pharmacology , Dioxygenases/antagonists & inhibitors , Dioxygenases/genetics , Flow Cytometry , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , Humans , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Temozolomide , Tumor Cells, Cultured , Tumor Stem Cell Assay
9.
Acta Neuropathol ; 122(4): 495-510, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21863242

ABSTRACT

Glioblastoma (GBM) is a highly aggressive brain tumour, where patients respond poorly to radiotherapy and exhibit dismal survival outcomes. The mechanisms of radioresistance are not completely understood. However, cancer cells with an immature stem-like phenotype are hypothesised to play a role in radioresistance. Since the progenitor marker neuron-glial-2 (NG2) has been shown to regulate several aspects of GBM progression in experimental systems, we hypothesised that its expression would influence the survival of GBM patients. Quantification of NG2 expression in 74 GBM biopsies from newly diagnosed and untreated patients revealed that 50% express high NG2 levels on tumour cells and associated vessels, being associated with significantly shorter survival. This effect was independent of age at diagnosis, treatment received and hypermethylation of the O(6)-methylguanine methyltransferase (MGMT) DNA repair gene promoter. NG2 was frequently co-expressed with nestin and vimentin but rarely with CD133 and the NG2 positive tumour cells harboured genetic aberrations typical for GBM. 2D proteomics of 11 randomly selected biopsies revealed upregulation of an antioxidant, peroxiredoxin-1 (PRDX-1), in the shortest surviving patients. Expression of PRDX-1 was associated with significantly reduced products of oxidative stress. Furthermore, NG2 expressing GBM cells showed resistance to ionising radiation (IR), rapidly recognised DNA damage and effectuated cell cycle checkpoint signalling. PRDX-1 knockdown transiently slowed tumour growth rates and sensitised them to IR in vivo. Our data establish NG2 as an important prognostic factor for GBM patient survival, by mediating resistance to radiotherapy through induction of ROS scavenging enzymes and preferential DNA damage signalling.


Subject(s)
Antigens/biosynthesis , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , DNA Damage/genetics , Glioblastoma/genetics , Glioblastoma/radiotherapy , Proteoglycans/biosynthesis , Stem Cells/metabolism , Aged , Antigens/genetics , Antigens/radiation effects , Biomarkers, Tumor/radiation effects , Brain Neoplasms/pathology , DNA Damage/radiation effects , Female , Glioblastoma/pathology , Humans , Male , Middle Aged , Predictive Value of Tests , Prognosis , Proteoglycans/genetics , Proteoglycans/radiation effects , Radiation Tolerance , Radiation, Ionizing , Stem Cells/pathology , Stem Cells/radiation effects , Survival Rate/trends
10.
Cancer Res ; 70(21): 8255-8, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20959482

ABSTRACT

The cancer stem cell hypothesis postulates that tumors arise from, and are maintained by, a small subpopulation of cancer stem cells. This concept has recently become increasingly controversial, following a series of conflicting results. The cell-surface epitope CD133 has been proposed as a brain cancer stem cell marker, whereas a growing number of studies clearly show a tumorigenic potential among CD133(-) cells as well. Diverging results suggest that assays for isolating cancer stem cells impose a selection bias on which cells are defined as cancer stem cells. Here, we highlight some recent developments, with an emphasis on reports that call for caution in the acceptance of the brain cancer stem cell hypothesis.


Subject(s)
Brain Neoplasms/pathology , Neoplastic Stem Cells/pathology , AC133 Antigen , Antigens, CD/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Glycoproteins/metabolism , Humans , Neoplastic Stem Cells/metabolism , Peptides/metabolism
11.
Cancer Res ; 70(11): 4274-9, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20460538

ABSTRACT

Although CD133 has been proposed as a marker for brain tumor-initiating cells, studies show that a tumorigenic potential exists among CD133(-) glioma cells as well. However, it is not established whether the ability of CD133(-) cells to form tumors is a property confined to a small subpopulation, rather than a common trait associated with most glioma cell types. Thus, we used lentiviral vectors expressing green fluorescent protein under lineage-specific promoters to identify CD133(-) glioma cells expressing Nestin, glial fibrillary acidic protein (GFAP), and neuron-specific enolase (NSE). Flow cytometry analysis showed the presence of CD133(-) subpopulations expressing these markers in glioma cell lines and in primary cultures from human glioblastoma (GBM) biopsies. Moreover, analysis of cell cycle distribution showed that subgroups expressing Nestin, GFAP, and NSE uniformly contained actively cycling cells, when cultured in serum-containing medium and stem cell medium. These subpopulations were fluorescence-activated cell sorted from CD133(-) U373 glioma cells and implanted intracerebrally in severe combined immunodeficient mice. Moreover, we implanted Nestin-, GFAP-, and NSE-positive glioma cells sorted from a human GBM biopsy, following removal of CD133-positive cells. All the CD133(-) subpopulations produced tumors, with no significant differences in survival or tumor take rates. However, there was a trend toward lower take rates for CD133(-) glioma subpopulations expressing GFAP and NSE. These findings suggest that the ability to form tumors may be a general trait associated with different glioma cell phenotypes, rather than a property limited to an exclusive subpopulation of glioma stem cells.


Subject(s)
Biomarkers, Tumor/biosynthesis , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , AC133 Antigen , Animals , Antigens, CD/biosynthesis , Brain Neoplasms/classification , Cell Growth Processes/physiology , Cell Line, Tumor , Genetic Vectors , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Glioblastoma/classification , Glycoproteins/biosynthesis , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Intermediate Filament Proteins/biosynthesis , Intermediate Filament Proteins/genetics , Lentivirus/genetics , Mice , Mice, SCID , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nestin , Peptides , Phosphopyruvate Hydratase/biosynthesis , Phosphopyruvate Hydratase/genetics
12.
Biotechniques ; 44(7): Piii-Pvi, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18533897

ABSTRACT

Array comparative genomic hybridization (aCGH) is a powerful tool to detect relative DNA copy number at a resolution limited only by the coverage of bacterial artificial chromosomes (BACs) used to print the genomic array. The amount of DNA needed to perform a reliable aCGH analysis has been a limiting factor, especially on minute tissue samples where limited DNA is available. Here we report a simple, highly sensitive and reliable aCGH method to analyze samples of no more than 1 ng genomic DNA. The speed and simplicity of the technique are ideal for studies on small clinical samples such as needle biopsies.


Subject(s)
DNA, Neoplasm/analysis , Gene Dosage , Nucleic Acid Amplification Techniques/methods , Nucleic Acid Hybridization/methods , Cell Line, Tumor , Humans , Oligonucleotide Array Sequence Analysis
13.
Int J Cancer ; 122(4): 761-8, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-17955491

ABSTRACT

CD133 is a cell surface marker expressed on progenitors of haematopoietic and endothelial cell lineages. Moreover, several studies have identified CD133 as a marker of brain tumor-initiating cells. In this study, human glioblastoma multiforme biopsies were engrafted intracerebrally into nude rats. The resulting tumors were serially passaged in vivo, and monitored by magnetic resonance imaging. CD133 expression was analyzed at various passages. Tumors initiated directly from the biopsies expressed little or no CD133, and showed no contrast enhancement suggesting an intact blood-brain barrier. During passaging, the tumors gradually displayed more contrast enhancement, increased angiogenesis and a shorter survival. Real-time qPCR and immunoblots showed that this was accompanied by increased CD133 expression. Primary biopsy spheroids and xenograft tumors were subsequently dissociated and flow sorted into CD133 negative and CD133 positive cell populations. Both populations incorporated BrdU in cell culture, and expressed the neural precursor marker nestin. Notably, CD133 negative cells derived from 6 different patients were tumorgenic when implanted into the rat brains. For 3 of these patients, analysis showed that the resulting tumors contained CD133 positive cells. In conclusion, we show that CD133 negative glioma cells are tumorgenic in nude rats, and that CD133 positive cells can be obtained from these tumors. Upon passaging of the tumors in vivo, CD133 expression is upregulated, coinciding with the onset of angiogenesis and a shorter survival. Thus, our findings do not suggest that CD133 expression is required for brain tumor initiation, but that it may be involved during brain tumor progression.


Subject(s)
Antigens, CD/metabolism , Brain Neoplasms/metabolism , Cell Proliferation , Glioblastoma/metabolism , Glycoproteins/metabolism , Peptides/metabolism , AC133 Antigen , Animals , Antigens, CD/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Disease Progression , Flow Cytometry , Glioblastoma/genetics , Glioblastoma/pathology , Glycoproteins/genetics , Humans , Immunoenzyme Techniques , Magnetic Resonance Imaging , Neovascularization, Pathologic , Peptides/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Nude , Tumor Cells, Cultured
14.
Curr Pharm Biotechnol ; 8(6): 320-5, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18289039

ABSTRACT

Emerging evidence suggests a class of non-coding RNAs termed microRNAs (miRNAs) play a key role in cancer. Since their original discovery in C. elegans in 1993 it has become evident that miRNAs are responsible for an entirely new mechanism of post-transcriptional gene regulation. miRNA expression is widespread in mammalian cells and notably altered in several cancer types. miRNA expression patterns correlate with several aspects of tumorigenesis and miRNA loci have been mapped to frequently altered cancer-associated genomic regions. Inhibition or augmentation of miRNA expression in cancer cells impacts gene expression and affects cell proliferation and survival. Hence, cancer-associated miRNAs may be regarded as a new class of non-coding tumour suppressors and oncogenes capable of regulating several key signalling pathways.


Subject(s)
MicroRNAs , Neoplasms/genetics , Animals , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/biosynthesis , MicroRNAs/genetics , Mutation , Neoplasms/therapy , Oncogenes
15.
Proc Natl Acad Sci U S A ; 103(44): 16466-71, 2006 Oct 31.
Article in English | MEDLINE | ID: mdl-17056721

ABSTRACT

In this work, highly infiltrative brain tumors with a stem-like phenotype were established by xenotransplantation of human brain tumors in immunodeficient nude rats. These tumors coopted the host vasculature and presented as an aggressive disease without signs of angiogenesis. The malignant cells expressed neural stem cell markers, showed a migratory behavior similar to normal human neural stem cells, and gave rise to tumors in vivo after regrafting. Serial passages in animals gradually transformed the tumors into an angiogenesis-dependent phenotype. This process was characterized by a reduction in stem cells markers. Gene expression profiling combined with high throughput immunoblotting analyses of the angiogenic and nonangiogenic tumors identified distinct signaling networks in the two phenotypes. Furthermore, proinvasive genes were up-regulated and angiogenesis signaling genes were down-regulated in the stem-like tumors. In contrast, proinvasive genes were down-regulated in the angiogenesis-dependent tumors derived from the stem-like tumors. The described angiogenesis-independent tumor growth and the uncoupling of invasion and angiogenesis, represented by the stem-like cancer cells and the cells derived from them, respectively, point at two completely independent mechanisms that drive tumor progression. This article underlines the need for developing therapies that specifically target the stem-like cell pools in tumors.


Subject(s)
Neoplasms/pathology , Stem Cells , Animals , Biopsy , Chromosomes, Human/genetics , Disease Progression , Gene Expression Regulation , Humans , Immunohistochemistry , Neoplasm Invasiveness , Neoplasm Transplantation , Neoplasms/blood supply , Neoplasms/genetics , Phenotype , Rats , Rats, Nude , Signal Transduction , Survival Rate , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...