Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(8)2020 Apr 22.
Article in English | MEDLINE | ID: mdl-32331440

ABSTRACT

Malignant gliomas are primary brain tumors with poor prognoses. These tumors are infiltrated by brain intrinsic microglia and peripheral monocytes which promote glioma cell invasion. In our previous studies, we discovered that the activation of Toll-like receptor 2 (TLR2) on microglia/brain macrophages converts them into a protumorigenic phenotype through the induction of matrix metalloproteinases (MMP) 9 and 14. In the present study, we used in vitro and in situ microglia-glioma interaction experimental models to test the impact of a novel inhibitor of TLR 2, ortho vanillin (O-Vanillin) to block TLR2 mediated microglia protumorigenic phenotype. We demonstrate that O-Vanillin inhibits the TLR2 mediated upregulation of MMP 9, MMP 14, IL 6 and iNOS expression. Similarly, the glioma supernatant induced MMP 9 and MMP 14 expression in murine and human microglia is abrogated by O-Vanillin treatment. O-Vanillin is not toxic for microglia, astrocytes or oligodendrocytes. Glioma growth in murine brain slice cultures is significantly reduced after treatment with O-Vanillin, and this reduced glioma growth depends on the presence of microglia. In addition, we also found that O-Vanillin inhibited the glioma induced proliferation of murine primary microglia. In summary, O-Vanillin attenuates the pro-tumorigenic phenotype of microglia/brain macrophages and thus qualifies as a candidate for glioma therapy.


Subject(s)
Benzaldehydes/pharmacology , Microglia/drug effects , Microglia/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Toll-Like Receptor 2/metabolism , Animals , Astrocytes/metabolism , Biomarkers , Cell Line, Tumor , Cell Proliferation , Cytokines/genetics , Cytokines/metabolism , Disease Susceptibility , Gene Expression Regulation , Glioma/etiology , Glioma/metabolism , Glioma/pathology , Humans , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Neoplasms/pathology , Signal Transduction
2.
Cell Rep ; 29(11): 3460-3471.e7, 2019 Dec 10.
Article in English | MEDLINE | ID: mdl-31825829

ABSTRACT

Microglia express Toll-like receptors (TLRs) that sense pathogen- and host-derived factors, including single-stranded RNA. In the brain, let-7 microRNA (miRNA) family members are abundantly expressed, and some have recently been shown to serve as TLR7 ligands. We investigated whether let-7 miRNA family members differentially control microglia biology in health and disease. We found that a subset of let-7 miRNA family members function as signaling molecules to induce microglial release of inflammatory cytokines, modulate antigen presentation, and attenuate cell migration in a TLR7-dependent manner. The capability of the let-7 miRNAs to control microglial function is sequence specific, mapping to a let-7 UUGU motif. In human and murine glioblastoma/glioma, let-7 miRNAs are differentially expressed and reduce murine GL261 glioma growth in the same sequence-specific fashion through microglial TLR7. Taken together, these data establish let-7 miRNAs as key TLR7 signaling activators that serve to regulate the diverse functions of microglia in health and glioma.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , MicroRNAs/metabolism , Microglia/metabolism , Toll-Like Receptor 7/genetics , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Glioma/pathology , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Signal Transduction , Toll-Like Receptor 7/metabolism
3.
Glia ; 64(8): 1416-36, 2016 08.
Article in English | MEDLINE | ID: mdl-27312099

ABSTRACT

Glioblastoma (GBM) is the most aggressive brain tumor in adults. It is strongly infiltrated by microglia and peripheral monocytes that support tumor growth. In the present study we used RNA sequencing to compare the expression profile of CD11b(+) human glioblastoma-associated microglia/monocytes (hGAMs) to CD11b(+) microglia isolated from non-tumor samples. Hierarchical clustering and principal component analysis showed a clear separation of the two sample groups and we identified 334 significantly regulated genes in hGAMs. In comparison to human control microglia hGAMs upregulated genes associated with mitotic cell cycle, cell migration, cell adhesion, and extracellular matrix organization. We validated the expression of several genes associated with extracellular matrix organization in samples of human control microglia, hGAMs, and the hGAMs-depleted fraction via qPCR. The comparison to murine GAMs (mGAMs) showed that both cell populations share a significant fraction of upregulated transcripts compared with their respective controls. These genes were mostly related to mitotic cell cycle. However, in contrast to murine cells, human GAMs did not upregulate genes associated to immune activation. Comparison of human and murine GAMs expression data to several data sets of in vitro-activated human macrophages and murine microglia showed that, in contrast to mGAMs, hGAMs share a smaller overlap to these data sets in general and in particular to cells activated by proinflammatory stimulation with LPS + INFγ or TNFα. Our findings provide new insights into the biology of human glioblastoma-associated microglia/monocytes and give detailed information about the validity of murine experimental models. GLIA 2016 GLIA 2016;64:1416-1436.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Microglia/metabolism , Monocytes/metabolism , Animals , CD11b Antigen/metabolism , Computational Biology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mice , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Transcriptome
4.
PLoS One ; 10(2): e0116644, 2015.
Article in English | MEDLINE | ID: mdl-25658639

ABSTRACT

Malignant glioma belong to the most aggressive neoplasms in humans with no successful treatment available. Patients suffering from glioblastoma multiforme (GBM), the highest-grade glioma, have an average survival time of only around one year after diagnosis. Both microglia and peripheral macrophages/monocytes accumulate within and around glioma, but fail to exert effective anti-tumor activity and even support tumor growth. Here we use microarray analysis to compare the expression profiles of glioma-associated microglia/macrophages and naive control cells. Samples were generated from CD11b+ MACS-isolated cells from naïve and GL261-implanted C57BL/6 mouse brains. Around 1000 genes were more than 2-fold up- or downregulated in glioma-associated microglia/macrophages when compared to control cells. A comparison with published data sets of M1, M2a,b,c-polarized macrophages revealed a gene expression pattern that has only partial overlap with any of the M1 or M2 gene expression patterns. Samples for the qRT-PCR validation of selected M1 and M2a,b,c-specific genes were generated from two different glioma mouse models and isolated by flow cytometry to distinguish between resident microglia and invading macrophages. We confirmed in both models the unique glioma-associated microglia/macrophage phenotype including a mixture of M1 and M2a,b,c-specific genes. To validate the expression of these genes in human we MACS-isolated CD11b+ microglia/macrophages from GBM, lower grade brain tumors and control specimens. Apart from the M1/M2 gene analysis, we demonstrate that the expression of Gpnmb and Spp1 is highly upregulated in both murine and human glioma-associated microglia/macrophages. High expression of these genes has been associated with poor prognosis in human GBM, as indicated by patient survival data linked to gene expression data. We also show that microglia/macrophages are the predominant source of these transcripts in murine and human GBM. Our findings provide new potential targets for future anti-glioma therapy.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Macrophages/physiology , Membrane Glycoproteins/genetics , Microglia/physiology , Osteopontin/genetics , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Cells, Cultured , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Humans , Macrophages/cytology , Membrane Glycoproteins/metabolism , Mice , Microglia/cytology , Neoplasm Transplantation , Osteopontin/metabolism , Prognosis
5.
J Cell Sci ; 127(Pt 17): 3687-98, 2014 09 01.
Article in English | MEDLINE | ID: mdl-24994934

ABSTRACT

The neuronal function of Cys-loop neurotransmitter receptors is established; however, their role in non-neuronal cells is poorly defined. As brain tumors are enriched in the neurotransmitter glycine, we studied the expression and function of glycine receptors (GlyRs) in glioma cells. Human brain tumor biopsies selectively expressed the GlyR α1 and α3 subunits, which have nuclear localization signals (NLSs). The mouse glioma cell line GL261 expressed GlyR α1, and knockdown of GlyR α1 protein expression impaired the self-renewal capacity and tumorigenicity of GL261 glioma cells, as shown by a neurosphere assay and GL261 cell inoculation in vivo, respectively. We furthermore showed that the pronounced tumorigenic effect of GlyR α1 relies on a new intracellular signaling function that depends on the NLS region in the large cytosolic loop and impacts on GL261 glioma cell gene regulation. Stable expression of GlyR α1 and α3 loops rescued the self-renewal capacity of GlyR α1 knockdown cells, which demonstrates their functional equivalence. The new intracellular signaling function identified here goes beyond the well-established role of GlyRs as neuronal ligand-gated ion channels and defines NLS-containing GlyRs as new potential targets for brain tumor therapies.


Subject(s)
Cytoplasm/metabolism , Glioma/metabolism , Receptors, Glycine/metabolism , Signal Transduction/physiology , Animals , Cell Line , Disease Models, Animal , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Glioma/pathology , Glycine/metabolism , Humans , Mice , Receptors, Glycine/genetics
6.
Int J Cancer ; 135(11): 2569-78, 2014 12 01.
Article in English | MEDLINE | ID: mdl-24752463

ABSTRACT

The invasiveness of malignant gliomas is one of the major obstacles in glioma therapy and the reason for the poor survival of patients. Glioma cells infiltrate into the brain parenchyma and thereby escape surgical resection. Glioma associated microglia/macrophages support glioma infiltration into the brain parenchyma by increased expression and activation of extracellular matrix degrading proteases such as matrix metalloprotease (MMP) 2, MMP9 and membrane-type 1 MMP. In this work we demonstrate that, MMP9 is predominantly expressed by glioma associated microglia/macrophages in mouse and human glioma tissue but not by the glioma cells. Supernatant from glioma cells induced the expression of MMP9 in cultured microglial cells. Using mice deficient for different Toll-like receptors we identified Toll-like receptor 2/6 as the signaling pathway for the glioma induced upregulation of microglial MMP9. Also in an experimental mouse glioma model, Toll-like receptor 2 deficiency attenuated the upregulation of microglial MMP9. Moreover, glioma supernatant triggered an upregulation of Toll-like receptor 2 expression in microglia. Both, the upregulation of MMP9 and Toll-like receptor 2 were attenuated by the antibiotic minocycline and a p38 mitogen-activated protein kinase antagonist in vitro. Minocycline also extended the survival rate of glioma bearing mice when given to the drinking water. Thus glioma cells change the phenotype of glioma associated microglia/macrophages in a complex fashion using Toll-like receptor 2 as an important signaling pathway and minocycline further proved to be a potential candidate for adjuvant glioma therapy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Brain Neoplasms/metabolism , Glioma/metabolism , Matrix Metalloproteinase 9/metabolism , Minocycline/pharmacology , Toll-Like Receptor 2/metabolism , Animals , Blotting, Western , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique , Glioma/drug therapy , Glioma/pathology , Humans , Immunoenzyme Techniques , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Toll-Like Receptor 2/genetics , Tumor Cells, Cultured
7.
Neuro Oncol ; 15(11): 1457-68, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24014382

ABSTRACT

BACKGROUND: Glioblastomas are the most aggressive primary brain tumors in humans. Microglia/brain macrophage accumulation in and around the tumor correlates with malignancy and poor clinical prognosis of these tumors. We have previously shown that microglia promote glioma expansion through upregulation of membrane type 1 matrix metalloprotease (MT1-MMP). This upregulation depends on signaling via the Toll-like receptor (TLR) adaptor molecule myeloid differentiation primary response gene 88 (MyD88). METHODS: Using in vitro, ex vivo, and in vivo techniques, we identified TLR2 as the main TLR controlling microglial MT1-MMP expression and promoting microglia-assisted glioma expansion. RESULTS: The implantation of mouse GL261 glioma cells into TLR2 knockout mice resulted in significantly smaller tumors, reduced MT1-MMP expression, and enhanced survival rates compared with wild-type control mice. Tumor expansion studied in organotypic brain slices depended on both parenchymal TLR2 expression and the presence of microglia. Glioma-derived soluble factors and synthetic TLR2 specific ligands induced MT1-MMP expression in microglia from wild-type mice, but no such change in MT1-MMP gene expression was observed in microglia from TLR2 knockout mice. We also found evidence that TLR1 and TLR6 cofunction with TLR2 as heterodimers in regulating MT1-MMP expression in vitro. CONCLUSIONS: Our results thus show that activation of TLR2 along with TLRs 1 and/or 6 converts microglia into a glioma supportive phenotype.


Subject(s)
Brain Neoplasms/metabolism , Brain/metabolism , Glioblastoma/metabolism , Macrophage Activation , Matrix Metalloproteinase 14/metabolism , Microglia/metabolism , Toll-Like Receptor 2/metabolism , Animals , Brain/pathology , Female , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Mice, Knockout , Survival Rate , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 6/metabolism
8.
Toxicol Pathol ; 40(8): 1169-87, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22659244

ABSTRACT

Smoking-associated chronic obstructive pulmonary disease is characterized by inflammation, changes affecting small airways, and development of emphysema. Various short- and long-term models have been introduced to investigate these processes. The aim of the present study was to identify markers of early epithelial injury/adaptation in a short-term animal model of cigarette smoke exposure. Initially, male BALB/c mice were exposed to smoke from one to five cigarettes and lung changes were assessed 4 and 24 hr after smoking cessation. Subsequently, animals were exposed to smoke from five cigarettes for 2 consecutive days and lungs investigated daily until the seventh postexposure day. Lung homogenates cytokines were determined, bronchioloalveolar fluid cells were counted, and lung tissue was analyzed by immunohistochemistry. Exposure to smoke from a single cigarette induced slight pulmonary neutrophilia. Smoke from two cigarettes additionally induced de novo expression of tight junction protein, claudin-3, by alveolar duct (AD) epithelial cells. Further increases in smoke exposure induced epithelial changes in airway progenitor regions. During the recovery period, the severity/frequency of epithelial reactions slowly decreased, coinciding with the switch from acute to a chronic inflammatory reaction. Claudin-3 and Clara cell 10 kDa protein were identified as possible markers of early tobacco smoke-induced epithelial injury along ADs.


Subject(s)
Alveolar Epithelial Cells/drug effects , Claudin-3/metabolism , Smoking/adverse effects , Uteroglobin/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Biomarkers/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Inhalation Exposure , Leukocyte Count , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Neutrophils/drug effects , Neutrophils/pathology , Time Factors , Nicotiana , Zonula Occludens-1 Protein/metabolism
9.
Reprod Toxicol ; 30(4): 613-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20955786

ABSTRACT

Antiepileptic drugs (AED) as transplacental agents are known to have adverse effects on fetal development. Genotoxicity of AEDs is still not fully understood. The aim of present study was to investigate the transplacental genotoxicity of valproate on animal model and in 21 mothers and their newborns receiving AED. In both studies, in vivo micronucleus (MN) assay was used. Pregnant dams were exposed to Na-valproate (100mg/kg) on gestational days 12-14. Dams and pups receiving Na-valproate showed a significantly increased MN frequency (5.17 ± 1.17/1000; 5.20 ± 1.48/1000) compared to the control (1.0 ± 0.58/1000; 1.67 ± 1.03/1000). In mother/newborn study a significant increase of MN frequency was detected in newborns of mothers taking AEDs (3.09 ± 0.49/10,000) compared to the referent newborns (1.56 ± 0.22/10,000). The results of this study suggest that AEDs may act as transplacental genotoxins. Launching the mother/newborn cohorts for genotoxicological monitoring may give a significant new insight in health effects of AEDs.


Subject(s)
Anticonvulsants/adverse effects , Anticonvulsants/toxicity , Epilepsy/drug therapy , Mutagens/adverse effects , Mutagens/toxicity , Pregnancy Complications/drug therapy , Prenatal Exposure Delayed Effects/blood , Adult , Animals , Animals, Newborn , Cohort Studies , Epilepsy/blood , Female , Humans , Infant, Newborn , Male , Mice , Mice, Inbred BALB C , Micronucleus Tests , Pilot Projects , Pregnancy , Pregnancy Complications/blood , Reticulocytes/drug effects , Valproic Acid/adverse effects , Valproic Acid/toxicity , Young Adult
10.
Mutat Res ; 657(1): 43-7, 2008 Nov 17.
Article in English | MEDLINE | ID: mdl-18789400

ABSTRACT

Over the last 40 years mankind has been facing new types of radiochemical environmental settings with every decade. During the last decade, biomonitoring was additionally focused on assessing associations between environmental exposure(s) and both early and late biological effects in children. Despite efforts to control and avoid child exposure to genotoxic agents the incidence of childhood cancers is increasing. Some cancers in adulthood may be the consequence of a multi-step process which starts with intrauterine and childhood exposure. This highlights the importance of a comprehensive interpretation of multiple health effects, especially considering recent studies suggesting that most health disorders are related to DNA changes. When exposed to genotoxic agents, a developing organism (fetus or child) is constantly being forced to reorganize into new equilibriums in order to adjust to a xenobiotic environment. In addition, the influence of sex hormones on radiochemical sensitivity is still unknown. For this reason special attention should be paid to puberty. The results of recent studies on animal models and follow up studies on children after nuclear accidents show long-lasting cytogenetic damage even after low dose exposures and their transgenerational persistance. To evaluate age-related difference and transplacental genotoxic potency fluconazole (FC) was investigated by in vivo micronucleus (MN) assay in adult mice, young mice and in transplacentally exposed newborn pups. Compared to the baseline values, FC caused no detectable genome damage in adult animals, but there was a significant increase in MN frequency in young animals and in newborn pups. Our study thus exemplifies an age-related chemosensitivity, and argues that cancer-promoting disturbances of complex prenatal developmental mechanisms and maturation during childhood require a new approach using systems biology.


Subject(s)
Aging , Fluconazole/toxicity , Maternal-Fetal Exchange , Mutagens/toxicity , Animals , Female , Mice , Mice, Inbred BALB C , Micronucleus Tests , Pregnancy
11.
Stem Cells ; 26(11): 2945-54, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18757298

ABSTRACT

Glioblastomas, the most aggressive primary brain tumors, occur almost exclusively in adult patients. Neural precursor cells (NPCs) are antitumorigenic in mice, as they can migrate to glioblastomas and induce tumor cell death. Here, we show that the antitumor effect of NPCs is age-dependently controlled by cell proliferation in the subventricular zone (SVZ) and that NPCs accumulating at a glioblastoma are diverted from their normal migratory path to the olfactory bulb. Experimentally induced cortical glioblastomas resulted in decreased subventricular proliferation in adult (postnatal day 90) but not in young (postnatal day 30) mice. Adult mice supplied fewer NPCs to glioblastomas and had larger tumors than young mice. Apart from the difference in proliferation, there was neither a change in cell number and death rate in the SVZ nor a change in angiogenesis and immune cell density in the tumors. The ability to kill glioblastomas was similar in NPCs isolated from young and adult mice. The proliferative response of NPCs to glioblastomas depended on the expression of D-type cyclins. In young mice, NPCs express the cyclins D1 and D2, but the expression of cyclin D1 is lost during aging, and in adult NPCs only cyclin D2 remains. In young and adult cyclin D2-deficient mice we observed a reduced supply of NPCs to glioblastomas and the generation of larger tumors compared with wild-type mice. We conclude that cyclin D1 and D2 are nonredundant for the antitumor response of subventricular NPCs. Loss of a single D-type cyclin results in a smaller pool of proliferating NPCs, lower number of NPCs migrating to the tumor, and reduced antitumor activity. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Neurons/transplantation , Stem Cells/cytology , Age Factors , Animals , Brain Neoplasms/pathology , Cell Proliferation , Cells, Cultured , Cyclin D1/metabolism , Cyclin D2 , Cyclins/metabolism , Glioblastoma/pathology , Mice , Mice, Inbred C57BL , Stem Cell Transplantation
12.
Brain ; 130(Pt 2): 476-89, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17107968

ABSTRACT

The invasion of tumour cells into brain tissue is a pathologic hallmark of WHO grades II-IV gliomas and contributes significantly to the failure of current therapeutic treatments. Activated microglial cells are abundant in brain tumours and may support tumour invasiveness. We have previously demonstrated that cyclosporin A (CsA) can affect growth of glioma cells in vitro by inhibiting signalling pathways, which are essential for tumour proliferation and invasiveness. In this work, we demonstrate that migration of EGFP-transfected glioblastoma cells in organotypic brain slices was significantly inhibited by treatment with CsA. On average 77% of untreated cells migrated beyond 500 mum, while only 28-33% cells migrated as far in the brain slices treated with CsA (P < 0.001). This inhibitory effect on glioblastoma invasion was lost when glioblastoma cells were injected into microglia-depleted brain slices. Moreover, CsA significantly inhibits intracranial glioma growth in vivo. We demonstrate that microglia-derived factors increase glioma invasiveness in Matrigel assay in vitro and this is associated with activation of the PI-3K/Akt signalling pathway. The invasion promoting effect of microglia is abolished in the presence of CsA. Furthermore, glioma-derived soluble factors induce morphological transformation of microglia and activate MAPK signalling, although production of pro-inflammatory factors was not observed. Our findings that CsA interferes at clinically relevant concentrations with the tumour-promoting role of microglia and impairs invasive growth of glioma cells in vivo may provide a novel therapeutic strategy against gliomas.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Cyclosporine/pharmacology , Glioblastoma/pathology , Microglia/drug effects , Animals , Antineoplastic Agents/therapeutic use , Brain Neoplasms/prevention & control , Cell Division/drug effects , Cell Movement/drug effects , Cyclosporine/therapeutic use , Glioblastoma/prevention & control , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Microglia/physiology , Neoplasm Invasiveness , Neoplasm Transplantation , Rats , Rats, Wistar , Signal Transduction/drug effects , Tissue Culture Techniques , Tumor Cells, Cultured
13.
Cancer Res ; 66(17): 8550-7, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951167

ABSTRACT

We report that experimental glioblastoma grow more vigorously in A(1) adenosine receptor (A(1)AR)-deficient mice associated with a strong accumulation of microglial cells at and around the tumors. A(1)ARs were prominently expressed in microglia associated with tumor cells as revealed with immunocytochemistry but low in microglia in the unaffected brain tissue. The A(1)AR could also be detected on microglia from human glioblastoma resections. To study functional interactions between tumor and host cells, we studied glioblastoma growth in organotypical brain slice cultures. A(1)AR agonists suppressed tumor growth. When, however, microglial cells were depleted from the slices, the agonists even stimulated tumor growth. Thus, adenosine attenuates glioblastoma growth acting via A(1)AR in microglia.


Subject(s)
Brain Neoplasms/pathology , Cell Communication/physiology , Glioblastoma/pathology , Microglia/cytology , Receptor, Adenosine A1/physiology , Adenosine/pharmacology , Animals , Cell Communication/drug effects , Cell Division , Cells, Cultured , Genes, Reporter , Genotype , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microglia/physiology , Receptor, Adenosine A1/deficiency , Receptor, Adenosine A1/genetics
14.
J Neuropathol Exp Neurol ; 64(9): 754-62, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16141784

ABSTRACT

Gliomas represent the most frequent type of human brain tumor, and their strong invasiveness is a significant clinical problem. Microglia, the immunocompetent cells of the brain, contribute significantly to the tumor and are potential interaction partners of the glioma cells. We studied the impact of the presence of microglia on tumor cell invasion in cultured brain slices. To selectively deplete microglia, the slices were treated with clodronate-filled liposomes. When glioma cells were injected into slices devoid of endogenous microglia, the invasiveness of the tumors was significantly decreased as compared with controls. Inoculation of exogenous microglia together with glioma cells into cultured brain slices increased the infiltrative behavior of the tumor depending on the microglia/glioma cell ratio. Cell culture experiments revealed that soluble factors released from glioma cells strongly stimulate metalloprotease-2 activity in microglia. In the brain slices inoculated with glioma cells, increased activity of metalloprotease-2 was directly correlated with the abundance of microglia. Our data indicate that glioma cells stimulate microglial cells to increase breakdown of extracellular matrix and thereby promote tumor invasiveness.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Metalloproteases/metabolism , Microglia/enzymology , Neoplasm Invasiveness/pathology , Animals , Antimetabolites/administration & dosage , Brain Neoplasms/metabolism , Cell Line, Tumor , Clodronic Acid/administration & dosage , Fluorescent Antibody Technique , Glioma/metabolism , Humans , Liposomes , Male , Mice , Microglia/drug effects , Microscopy, Confocal , Organ Culture Techniques
15.
Environ Mol Mutagen ; 46(1): 59-63, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15880513

ABSTRACT

The antibiotic 5-nitrofurantoin (5-NF) has been used widely for the treatment of urosepsis in children during the last 20 years. Recent experimentation suggests the need for reevaluating its genotoxic potential. Because of possible differences in the metabolism and clearance of 5-NF in young and adult animals, we conducted a study to determine whether micronuclei caused by 5-NF were age-related. The in vivo micronucleus (MN) assay was performed on 3- and 8-week-old mice given single intraperitoneal injections of 5, 10, and 50 mg/kg 5-NF. Blood samples from the tail vein were taken before injection (baseline) and at 48, 96, 168, and 336 hr (2 weeks) after the treatment. One thousand reticulocytes were analyzed for micronuclei from each animal. Compared to similar baseline values for young and adult mice, 5-NF caused a significant increase in MN frequency in both age groups. The mean MN frequency in the young animals was higher than in the adult animals for each dose and sampling time. MN frequencies remained significantly elevated in young animals even 2 weeks after exposure to 5-NF. The results of the study confirm the genotoxic potential of 5-NF in young and adult animals, and indicate that young animals are more sensitive to the genotoxic effects of 5-NF than adult mice and that the response in young mice persists for a significantly longer time. These findings may be related to poorly developed mechanisms of xenobiotic detoxification and renal elimination in young animals.


Subject(s)
Aging/metabolism , Anti-Infective Agents, Urinary/adverse effects , Micronuclei, Chromosome-Defective/chemically induced , Nitrofurantoin/adverse effects , Analysis of Variance , Animals , Anti-Infective Agents, Urinary/pharmacokinetics , Female , Inactivation, Metabolic , Male , Mice , Mice, Inbred BALB C , Micronucleus Tests , Nitrofurantoin/pharmacokinetics , Reticulocytes/drug effects , Time Factors
16.
J Neurosci ; 25(10): 2637-46, 2005 Mar 09.
Article in English | MEDLINE | ID: mdl-15758174

ABSTRACT

Neural precursor cells contribute to adult neurogenesis and to limited attempts of brain repair after injury. Here we report that in a murine experimental glioblastoma model, endogenous neural precursors migrate from the subventricular zone toward the tumor and surround it. The association of endogenous precursors with syngenic tumor grafts was observed, after injecting red fluorescent protein-labeled G261 cells into the caudate-putamen of transgenic mice, which express green fluorescent protein under a promoter for nestin (nestin-GFP). Fourteen days after inoculation, the nestin-GFP cells surrounded the tumors in several cell layers and expressed markers of early noncommitted and committed precursors. Nestin-GFP cells were further identified by a characteristic membrane current pattern as recorded in acute brain slices. 5-bromo-2-deoxyuridine labeling and dye tracing experiments revealed that the tumor-associated precursors originated from the subventricular zone. Moreover, in cultured explants from the subventricular zone, the neural precursors showed extensive tropism for glioblastomas. Tumor-induced endogenous precursor cell accumulation decreased with age of the recipient; this correlated with increased tumor size and shorter survival times in aged mice. Coinjection of glioblastoma cells with neural precursors improved the survival time of old mice to a level similar to that in young mice. Coculture experiments showed that neural precursors suppressed the rapid increase in tumor cell number, which is characteristic of glioblastoma, and induced glioblastoma cell apoptosis. Our results indicate that tumor cells attract endogenous precursor cells; the presence of precursor cells is antitumorigenic; and this cellular interaction decreases with aging.


Subject(s)
Cell Communication , Cell Movement/physiology , Glioblastoma/mortality , Glioblastoma/pathology , Neurons/cytology , Neurons/transplantation , Stem Cells/cytology , Animals , Caudate Nucleus/cytology , Caudate Nucleus/transplantation , Cell Communication/physiology , Cell Line, Tumor , Cells, Cultured , Coculture Techniques/methods , Glioblastoma/surgery , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rats , Stem Cell Transplantation/methods , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...