Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Pathol ; 224(2): 222-33, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21462187

ABSTRACT

Matrix metalloproteinase 2 (MMP2) is an extracellular protein-degrading enzyme widely believed to be involved in the invasion of brain tumour cells. However, this assumption is mainly based on in vitro studies. By characterizing the transcriptome and in vivo properties of 20 astrocytoma cell lines, we found that the levels of MMP2 were higher in GFAP(-) astrocytoma cells and correlated with their ability to induce vascular changes, a common complication of malignant tumours. To study the relationship between MMP2 expression and vascular alteration, we intracerebrally implanted immunodeficient mice with human astrocytoma cells stably transduced with lentiviral vectors expressing either MMP2 or a short hairpin RNA against MMP2. We found that the tumours depleted of MMP2 were larger, contained more proliferating cells and fewer macrophages, and had a vasculature that was more destabilized and regressed with fewer capillary sprouts. In contrast, the tumours overexpressing MMP2 were smaller and showed no histological difference compared to the controls. We therefore suggest that MMP2 is not the cause of vascular atypia in malignant brain tumours, but is involved in a tissue repair response that tends to limit the growth of these tumours. This study argues against MMP2 inhibition as a therapeutic approach for brain cancer and provides a comprehensive characterization of popular astrocytoma cell lines that should help to identify alternative targets.


Subject(s)
Astrocytoma/pathology , Brain Neoplasms/pathology , Macrophages/pathology , Matrix Metalloproteinase 2/physiology , Neovascularization, Pathologic/pathology , Animals , Astrocytoma/blood supply , Astrocytoma/enzymology , Brain Neoplasms/blood supply , Brain Neoplasms/enzymology , Gene Knockdown Techniques , Genetic Vectors , Humans , Lentivirus/genetics , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, SCID , Neoplasm Transplantation , Neovascularization, Pathologic/enzymology , Transplantation, Heterologous , Tumor Cells, Cultured
2.
Brain Pathol ; 21(6): 661-71, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21418369

ABSTRACT

The cerebral vasculature is constantly patrolled by rod-shaped leukocytes crawling on the luminal endothelial surface. These cells are recruited in greater numbers after exposure to bacterial lipopolysaccharide (LPS) by a mechanism involving tumor necrosis factor (TNF), interleukin-1ß (IL1ß) and angiopoietin-2 (Angpt2). Here, we report that the population of crawling leukocytes, consisting mainly of granulocytes, is also increased in the brains of mice suffering from experimental autoimmune encephalomyelitis (EAE) or injected with pertussis toxin (PTX), which is commonly used to induce EAE. However, this recruitment occurs through an alternative mechanism, independent of Angpt2. In a series of experiments using DNA microarrays, knockout mice and neutralizing antibodies, we found that PTX acts indirectly on the endothelium in part through IL6, which is essential for the post-transcriptional upregulation of intercellular adhesion molecule 1 (ICAM1) in response to PTX but not to LPS. We also found that phagocytes adhere to brain capillaries through the interaction of integrin αM (ITGαM) with ICAM1 and an unidentified ligand. In conclusion, this study supports the concept that PTX promotes EAE, at least in part, by inducing vascular changes necessary for the recruitment of patrolling leukocytes.


Subject(s)
Adjuvants, Immunologic/pharmacology , Brain/immunology , Chemotaxis, Leukocyte/immunology , Granulocytes/immunology , Pertussis Toxin/pharmacology , Phagocytes/immunology , Animals , Brain/blood supply , Brain/pathology , CD11b Antigen/immunology , CD11b Antigen/metabolism , Cell Adhesion/immunology , Cell Separation , Chemotaxis, Leukocyte/drug effects , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Granulocytes/metabolism , In Situ Hybridization , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , Phagocytes/metabolism , Polymerase Chain Reaction
3.
Wound Repair Regen ; 19(1): 38-48, 2011.
Article in English | MEDLINE | ID: mdl-21143691

ABSTRACT

The anterior cruciate ligament (ACL) is often the target of knee trauma. This ligament does not heal very well, leading to joint instability. Long-term instability of the knee can lead to early arthritis and loss of function. To develop efficient strategies to stimulate posttraumatic ACL regeneration in vivo, a good healing model is needed in vitro. Such a model must remain as simple as possible, but should include key features to provide relevant answers to precise questions about the clinical problem addressed. Here, we report tissue-engineered type I collagen scaffolds developed to establish an ACL healing model in vitro and a potential ACL substitute in vivo. Such scaffolds were used to evaluate ACL cell growth, migration, and the capacity to synthesize and assemble collagen fibers for up to 40 days in vitro and up to 180 days in vivo. They were anchored with two bone plugs to allow their static stretching in culture and to facilitate their surgical implantation in knee joints. Our results have shown that living ACL fibroblasts can attach, migrate, and colonize this type of scaffold. In vitro, the cells populated the scaffolds and expressed mRNAs coding for the prolyl-4-hydroxylase, involved in collagen fibers' assembly. In vivo, acellular implants were vascularized and populated with caprine cells that migrated from the osseous insertions toward the center of the grafts. This model is a very good tool to study ACL repair and identify the factors that could accelerate its healing postsurgery.


Subject(s)
Anterior Cruciate Ligament/physiology , Collagen Type I/physiology , Fibroblasts/physiology , Tissue Engineering/methods , Tissue Scaffolds , Wound Healing/physiology , Animals , Goats , Humans , Knee Injuries/therapy , Models, Biological , Tissue Culture Techniques
4.
Cell Transplant ; 20(4): 535-42, 2011.
Article in English | MEDLINE | ID: mdl-21054949

ABSTRACT

Fibroblasts isolated from skin and from anterior cruciate ligament (ACL) secrete type I and type III collagens in vivo and in vitro. However, it is much easier and practical to obtain a small skin biopsy than an ACL sample to isolate fibroblasts for tissue engineering applications. Various tissue engineering strategies have been proposed for torn ACL replacement. We report here the results of the implantation of bioengineered ACLs (bACLs), reconstructed in vitro using a type I collagen scaffold, anchored with two porous bone plugs to allow bone-ligament-bone surgical engraftment. The bACLs were seeded with autologous living dermal fibroblasts, and grafted for 6 months in goat knee joints. Histological and ultrastructural observations ex vivo demonstrated a highly organized ligamentous structure, rich in type I collagen fibers and cells. Grafts' vascularization and innervation were observed in all bACLs that were entirely reconstructed in vitro. Organized Sharpey's fibers and fibrocartilage, including chondrocytes, were present at the osseous insertion sites of the grafts. They showed remodeling and matrix synthesis postimplantation. Our tissue engineering approach may eventually provide a new solution to replace torn ACL in humans.


Subject(s)
Anterior Cruciate Ligament/cytology , Anterior Cruciate Ligament/transplantation , Fibroblasts/cytology , Tissue Engineering/methods , Animals , Cattle , Cells, Cultured , Collagen Type I/chemistry , Collagen Type I/metabolism , Collagen Type III/chemistry , Collagen Type III/metabolism , Female , Goats , Humans
5.
Brain Pathol ; 18(3): 401-14, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18371178

ABSTRACT

All patients with glioblastoma, the most aggressive and common form of brain cancer, develop cerebral edema. This complication is routinely treated with dexamethasone, a steroidal anti-inflammatory drug whose effects on brain tumors are not fully understood. Here we show that dexamethasone can reduce glioma growth in mice, even though it depletes infiltrating T cells with potential antitumor activity. More precisely, T cells with helper or cytotoxic function were sensitive to dexamethasone, but not those that were negative for the CD4 and CD8 molecules, including gammadelta and natural killer (NK) T cells. The antineoplastic effect of dexamethasone was indirect, as it did not meaningfully affect the growth and gene expression profile of glioma cells in vitro. In contrast, hundreds of dexamethasone-modulated genes, notably angiopoietin 2 (Angpt2), were identified in cultured cerebral endothelial cells by microarray analysis. The ability of dexamethasone to attenuate Angpt2 expression was confirmed in vitro and in vivo. Selective neutralization of Angpt2 using a peptide-Fc fusion protein reduced glioma growth and vascular enlargement to a greater extent than dexamethasone, without affecting T cell infiltration. In conclusion, this study suggests a mechanism by which dexamethasone can slow glioma growth, providing a new therapeutic target for malignant brain tumors.


Subject(s)
Angiopoietin-2/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Brain Neoplasms/drug therapy , Dexamethasone/pharmacology , Glioma/drug therapy , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Flow Cytometry , Gene Expression/drug effects , Glioma/blood supply , Glioma/metabolism , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Oligonucleotide Array Sequence Analysis , Recombinant Fusion Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/drug effects
6.
Glia ; 55(8): 790-800, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17390309

ABSTRACT

G protein-coupled receptor 84 (GPR84) is a recently discovered member of the seven transmembrane receptor superfamily whose function and regulation are unknown. Here, we report that in mice suffering from endotoxemia, microglia express GPR84 in a strong and sustained manner. This property is shared by subpopulations of peripheral macrophages and, to a much lesser extent, monocytes. The induction of GPR84 expression by endotoxin is mediated, at least in part, by proinflammatory cytokines, notably tumor necrosis factor (TNF) and interleukin-1 (IL-1), because mice lacking either one or both of these molecules have fewer GPR84-expressing cells in their cerebral cortex than wild-type mice during the early phase of endotoxemia. Moreover, when injected intracerebrally or added to microglial cultures, recombinant TNF stimulates GPR84 expression through a dexamethasone-insensitive mechanism. Finally, we show that microglia produce GPR84 not only during endotoxemia, but also during experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis. In conclusion, this study reports the identification of a new sensitive marker of microglial activation, which may play an important regulatory role in neuroimmunological processes, acting downstream to the effects of proinflammatory mediators.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Endotoxemia/immunology , Microglia/immunology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Animals , Brain/immunology , Cells, Cultured , Dexamethasone/pharmacology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Endotoxemia/physiopathology , Gene Expression/drug effects , Gene Expression/immunology , Glucocorticoids/pharmacology , Interleukin-1/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/drug effects , Monocytes/immunology , Tumor Necrosis Factor-alpha/pharmacology
7.
Glia ; 55(8): 777-89, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17285589

ABSTRACT

Microglia, monocytes, and peripheral macrophages share a common origin and many characteristics, but what distinguishes them from each other at the level of gene expression remains largely unknown. In this study, we compared the transcriptional profiles of freshly purified microglia, monocytes, and spleen macrophages using Affymetrix Mouse Genome arrays to identify genes predominantly expressed by microglia. Among tens of thousands of genes assayed, 127 potential candidates were found, including nine newly discovered genes encoding plasma membrane and extracellular proteins. In the brain, the latter were selectively expressed by microglia, as revealed by in situ hybridization. Three of them were confirmed to be exclusively (MSR2) or predominantly (GPR12, GPR34) expressed in the brain compared to the other tissues examined. Furthermore, all of these genes were upregulated in activated microglia after treatment with the demyelinating toxin cuprizone, suggesting that they play roles in neuroinflammation. In conclusion, this study reports the identification of new selective markers for microglia, which should prove useful not only to identify and isolate these cells, but also to better understand their distinctive properties.


Subject(s)
Biomarkers , Microglia/immunology , Neuritis/genetics , Neuritis/immunology , Oligonucleotide Array Sequence Analysis , Animals , Brain/cytology , Brain/immunology , Chelating Agents/toxicity , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/genetics , Demyelinating Diseases/immunology , Disease Models, Animal , Gene Expression/drug effects , Gene Expression/immunology , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Neuritis/chemically induced , RNA, Messenger/analysis , Spleen/cytology , Spleen/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
8.
Cancer Res ; 65(9): 3928-36, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15867393

ABSTRACT

Recent findings implicate macrophages and some of their secreted products, especially tumor necrosis factor (TNF), as tumor promoters. Inhibitors of these inflammatory components are currently regarded as potential therapeutic tools to block tumor progression. Here, we show that infiltrating macrophages represented a significant population of nonneoplastic cells within malignant gliomas, in which they were the exclusive producers of TNF. Contrary to the reported pro-oncogenic effects of TNF in other types of solid tumors, glioma-bearing mice deficient in TNF developed larger tumors and had reduced survival compared with their wild-type controls. Histologic examinations revealed that glioma volume was negatively correlated with the number of macrophages and small cavities called microcysts. Overall, our results support the concept that macrophages alter brain tumor development through a TNF-dependent process that culminates in the formation of microcysts. This raises the question of whether anti-inflammatory drugs, such as those commonly administrated to patients with brain cancer, could interfere with antitumor mechanisms.


Subject(s)
Brain Neoplasms/immunology , Glioma/immunology , Macrophages/immunology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/immunology , Animals , Brain Neoplasms/pathology , Cell Growth Processes/immunology , Chickens , Glioma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology
9.
Eur Cell Mater ; 7: 1-11; discussion 1-11, 2004 Mar 10.
Article in English | MEDLINE | ID: mdl-15015133

ABSTRACT

The isolation of human bronchial epithelial (HBEC) and fibroblastic cells (HBFC) from biopsies of asthmatic and non-asthmatic volunteers provided unique cellular materials to be used for the production of bioengineered bronchial equivalents (BE) in vitro. The HBEC are grown on a mesenchymal layer seeded with HBFC and the BE can be maintained for at least 15 days in culture. Under the BE culture conditions established previously, HBEC undergo differentiation into ciliated and goblet cells, within a pseudostratified organization comparable to human bronchi. We published previously the results from histologic and functional analyses of such BE produced exclusively using non-asthmatic HBEC and HBFC. We report here the comparative analyses of BE produced with non-asthmatic and asthmatic living HBEC and HBFC (naBE and aBE, respectively). Our data indicated that all asthmatic HBEC populations grown on a mesenchymal layer, containing non-asthmatic HBFC, slowly reached a confluent state but then detached from the matrix upon culture time. These BE appear to be very good models to study the mechanisms involved in asthma in vitro.


Subject(s)
Asthma/pathology , Bronchi/cytology , Epithelial Cells/cytology , Tissue Engineering , Adult , Animals , Biopsy , Bronchi/pathology , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Cilia/ultrastructure , Culture Media/pharmacology , Culture Media, Conditioned/pharmacology , Culture Techniques/methods , Dogs , Epithelial Cells/enzymology , Fibroblasts/cytology , Gelatinases/metabolism , Humans , Mesoderm , Microscopy, Electron , Middle Aged , Rats , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...