Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancer Med ; 3(4): 812-24, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24810477

ABSTRACT

It is well known that in vitro subculture represents a selection pressure on cell lines, and over time this may result in a genetic drift in the cancer cells. In addition, long-term cultures harbor the risk of cross-contamination with other cell lines. The consequences may have major impact on experimental results obtained in various laboratories, where the cell lines no longer reflect the original tumors that they are supposed to represent. Much neglected in the scientific community is a close monitoring of cell cultures by regular phenotypic and genetic characterization. In this report, we present a thorough characterization of the commonly used glioblastoma (GBM) model U-251, which in numerous publications has been wrongly identified as U-373, due to an earlier cross-contamination. In this work, the original U-251 and three subclones of U-251, commonly referred to as U-251 or U-373, were analyzed with regard to their DNA profile, morphology, phenotypic expression, and growth pattern. By array comparative genomic hybridization (aCGH), we show that only the original low-passaged U-251 cells, established in the 1960s, maintain a DNA copy number resembling a typical GBM profile, whereas all long-term subclones lost the typical GBM profile. Also the long-term passaged subclones displayed variations in phenotypic marker expression and showed an increased growth rate in vitro and a more aggressive growth in vivo. Taken together, the variations in genotype and phenotype as well as differences in growth characteristics may explain different results reported in various laboratories related to the U-251 cell line.


Subject(s)
Cell Line, Tumor/physiology , Glioblastoma/pathology , Animals , Carcinogenesis , Cell Proliferation , Cell Shape , Comparative Genomic Hybridization , Female , Gene Expression , Genetic Drift , Humans , Karyotype , Male , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Phenotype , Ploidies
2.
PLoS One ; 6(7): e23062, 2011.
Article in English | MEDLINE | ID: mdl-21829586

ABSTRACT

Aberrant expression of the progenitor marker Neuron-glia 2 (NG2/CSPG4) or melanoma proteoglycan on cancer cells and angiogenic vasculature is associated with an aggressive disease course in several malignancies including glioblastoma multiforme (GBM) and melanoma. Thus, we investigated the mechanism of NG2 mediated malignant progression and its potential as a therapeutic target in clinically relevant GBM and melanoma animal models. Xenografting NG2 overexpressing GBM cell lines resulted in increased growth rate, angiogenesis and vascular permeability compared to control, NG2 negative tumours. The effect of abrogating NG2 function was investigated after intracerebral delivery of lentivirally encoded shRNAs targeting NG2 in patient GBM xenografts as well as in established subcutaneous A375 melanoma tumours. NG2 knockdown reduced melanoma proliferation and increased apoptosis and necrosis. Targeting NG2 in two heterogeneous GBM xenografts significantly reduced tumour growth and oedema levels, angiogenesis and normalised vascular function. Vascular normalisation resulted in increased tumour invasion and decreased apoptosis and necrosis. We conclude that NG2 promotes tumour progression by multiple mechanisms and represents an amenable target for cancer molecular therapy.


Subject(s)
Antigens/metabolism , Brain Neoplasms/prevention & control , Cell Proliferation , Disease Models, Animal , Glioblastoma/prevention & control , Melanoma/prevention & control , Neovascularization, Pathologic , Proteoglycans/antagonists & inhibitors , Proteoglycans/metabolism , Animals , Antigens/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Adhesion , Cell Movement , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunoenzyme Techniques , Magnetic Resonance Imaging , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Proteoglycans/genetics , RNA, Small Interfering/genetics , Rats , Rats, Nude , Transgenes/physiology
3.
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
5.
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
6.
Cancer Res ; 69(13): 5331-9, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19509230

ABSTRACT

Human mesenchymal stem cells (hMSC) aid in tissue maintenance and repair by differentiating into specialized cell types. Due to this ability, hMSC are currently being evaluated for cell-based therapies of tissue injury and degenerative diseases. However, extensive expansion ex vivo is a prerequisite to obtain the cell numbers required for human cell-based therapy protocols. Recent studies indicate that hMSC may contribute to cancer development and progression either by acting as cancer-initiating cells or through interactions with stromal elements. If spontaneous transformation ex vivo occurs, this may jeopardize the use of hMSC as therapeutic tools. Whereas murine MSC readily undergo spontaneous transformation, there are conflicting reports about spontaneous transformation of hMSC. We have addressed this controversy in a two-center study by growing bone marrow-derived hMSC in long-term cultures (5-106 weeks). We report for the first time spontaneous malignant transformation to occur in 45.8% (11 of 24) of these cultures. In comparison with hMSC, the transformed mesenchymal cells (TMC) showed a significantly increased proliferation rate and altered morphology and phenotype. In contrast to hMSC, TMC grew well in soft agar assays and were unable to undergo complete differentiation. Importantly, TMC were highly tumorigenic, causing multiple fast-growing lung deposits when injected into immunodeficient mice. We conclude that spontaneous malignant transformation may represent a biohazard in long-term ex vivo expansion of hMSC. On the other hand, this spontaneous transformation process may represent a unique model for studying molecular pathways initiating malignant transformation of hMSC.


Subject(s)
Bone Marrow Cells/cytology , Cell Transformation, Neoplastic , Mesenchymal Stem Cells/cytology , Adolescent , Adult , Animals , Bone Marrow Cells/pathology , Cell Culture Techniques/methods , Cell Differentiation , Cell Division , Flow Cytometry , Humans , Kinetics , Mesenchymal Stem Cells/pathology , Mice , Middle Aged , Phenotype , Reference Values , Species Specificity , Time Factors , Young Adult
7.
FASEB J ; 22(9): 3120-8, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18495755

ABSTRACT

A NOD/Scid mouse expressing enhanced green fluorescent protein (eGFP) is described, in which human and mouse tumors marked with red fluorescent protein can be established in vivo, both at subcutaneous and orthotopic locations. Using light microscopy as well as multiphoton confocal microscopy techniques, we visualized in detail the intricate colocalization of tumor and host cells in situ. Moreover, using fluorescence-activated cell sorting (FACS), we were able to completely separate the host cells from the tumor cells, thus providing a system for detailed cellular and molecular analysis of tumor-host cell interactions. The fact that tumor and host cells can be reliably identified also allowed us to detect double-positive cells, possibly arising from cell fusion events or horizontal gene transfer. Similarly, the model can be applied for the detection of circulating metastatic cells and for detailed studies on the vascular compartments within tumors, including vasculogenic mimicry. Thus, the model described should provide significant insight into how tumor cells communicate with their microenvironment.


Subject(s)
Green Fluorescent Proteins/biosynthesis , Mice, SCID , Neoplasms/physiopathology , Animals , Cell Communication/physiology , Humans , Mice , Mice, Inbred NOD , Microscopy, Fluorescence, Multiphoton , Neoplasm Transplantation/immunology , Neoplasm Transplantation/pathology , Neoplasms/immunology , Neoplasms/pathology
8.
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
9.
J Gene Med ; 8(9): 1131-40, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16810631

ABSTRACT

BACKGROUND: Adeno-associated viral (AAV) vectors are potent delivery vehicles for gene transfer strategies directed at the central nervous system (CNS), muscle and liver. However, comparatively few studies have described AAV-mediated gene transfer to tumor tissues. We have previously demonstrated that while AAV2 and Adenoviral (Ad) 5 vectors have similar broad host ranges in tumor-derived cell lines, AAV2 was able to penetrate human glioblastoma biopsy spheroids and xenografts more efficiently than Ad 5 vectors. These results suggested that AAV vectors could be suitable for therapeutic gene delivery to solid tumor tissue. In the present work, the transduction efficacy of AAV serotypes 4 and 5 were compared to AAV2, both in vitro and in intracranial GBM xenografts derived from patient biopsies implanted into nude rats. METHODS: AAV vector serotypes 2, 4, and 5 containing either the green fluorescent protein (GFP) or the bacterial beta-galactosidase (lacZ) reporter gene were added to five different human glioma cell lines, to multicellular spheroids generated from glioblastoma patient biopsies, and to spheroids xenografted intracranially in nude rats. Transduction efficiency was assessed by fluorescence imaging, histochemistry, immunohistochemistry and flow cytometry. RESULTS: While all three AAV serotypes were able to transduce the glioma cell lines when added individually or when they were administered in concert, AAV2 transduced the glioma cells most effectively compared to AAV4 or AAV5. Upon infecting glioblastoma spheroids in vitro, all three AAV serotypes efficiently transduced cells located at the surface as well as within deeper layers of the spheroids. In addition, similarly to what was observed for AAV2 16, both AAV4 and AAV5 were able to transduce human glioblastoma xenografts implanted intracranially. CONCLUSIONS: In addition to the widely used AAV2 serotype, AAV4 and AAV5 serotypes may also be used to transduce biologically diverse glioma cell lines. They also penetrate and transduce solid human tumor tissue derived from patient biopsies. Therefore, the data presented here provide a proof of principle for developing AAV4 and AAV5 as treatment vehicles for human malignant gliomas.


Subject(s)
Dependovirus/classification , Dependovirus/genetics , Genetic Therapy/methods , Glioma/therapy , Animals , Cell Line, Tumor , Female , Genes, Reporter , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/therapy , Glioma/genetics , Glioma/pathology , Green Fluorescent Proteins/genetics , Humans , Lac Operon , Male , Neoplasm Transplantation , Rats , Rats, Nude , Serotyping , Spheroids, Cellular , Transduction, Genetic , Transplantation, Heterologous
10.
Hum Gene Ther ; 16(3): 381-92, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15812233

ABSTRACT

The transduction patterns of recombinant adeno-associated virus serotype 1 (AAV1) and serotype 6 (AAV6) vectors were assessed in human glioblastoma multiforme (GBM) cell lines, in human GBM biopsy spheroids, and in tumor xenografts growing in nude rat brains. All the cell lines tested (A172, D37, GaMg, HF66, and U373Mg) were found to be permissive to both AAV1 and AAV6 vectors, and thus displayed a transduction pattern similar to AAV2 vectors. For every cell line tested, the transduction efficiency displayed by AAV2 vectors was better than by isogenic and isopromoter AAV1 vectors. Transduction efficiency was dependent on the viral particle number used, suggesting that the receptors for these vectors are widely distributed in GBM tissues. Interestingly, AAV1, AAV2, and AAV6 vectors were able to infect and transduce the same cells when added simultaneously to monolayer cultures. Infection of human GBM biopsy spheroids with AAV1 and AAV6 vectors resulted in transgene expression both at the surface layers and in the core of the spheroids. Following injection of AAV1 and AAV6 vectors into human GBM biopsy xenografts growing in nude rat brains, reporter gene expression was seen both in the periphery as well as in the central regions of the tumors. When injected into the normal rat brain, both AAV1 and AAV6 vectors were found to transduce several central nervous system (CNS) regions. The presented results suggest a potential therapeutic role for AAV1 and AAV6 vectors in gene therapy for GBM and also for other CNS malignancies.


Subject(s)
Brain Neoplasms/therapy , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/pharmacokinetics , Glioblastoma/therapy , Transduction, Genetic , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Genetic Vectors/genetics , Glioblastoma/metabolism , Histocytochemistry , Humans , Immunohistochemistry , Rats , Rats, Nude , Spheroids, Cellular/metabolism , Spheroids, Cellular/virology , Transplantation, Heterologous , beta-Galactosidase
SELECTION OF CITATIONS
SEARCH DETAIL
...