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.
Neuro Oncol ; 18(7): 939-49, 2016 07.
Article in English | MEDLINE | ID: mdl-26951383

ABSTRACT

BACKGROUND: Glioblastomas are highly vascularized tumors with a prominent infiltration of macrophages/microglia whose role in promoting glioma growth, invasion, and angiogenesis has not been fully elucidated. METHODS: The contribution of myeloid-derived vascular endothelial growth factor (VEGF) to glioma growth was analyzed in vivo in a syngeneic intracranial GL261 glioma model using a Cre/loxP system to knock out the expression of VEGF-A in CD11b + myeloid cells. Changes in angiogenesis-related gene expression profile were analyzed in mutant bone marrow-derived (BMD) macrophages in vitro. Furthermore, we studied the influence of macrophages on GL261 growth, invasiveness, and protein expression profile of angiogenic molecules as well as the paracrine effect of mutant macrophages on angiogenesis in vitro. RESULTS: Myeloid cell-restricted VEGF-A deficiency leads to a growth delay of intracranial tumors and prolonged survival. The tumor vasculature in mutant mice was more regular, with increased pericyte coverage. Expression analysis revealed significant downregulation of VEGF-A and slight upregulation of TGFß-1 in BMD macrophages from mutant mice. Endothelial tube formation was significantly decreased by conditioned media from mutant macrophages. The expression of angiogenesis-related proteins in GL261 glioma cells in co-culture experiments either with wild-type or mutant macrophages remained unchanged, indicating that effects observed in vivo are due to myeloid-derived VEGF-A deficiency. CONCLUSIONS: Our results highlight the importance of VEGF derived from tumor-infiltrating myeloid cells for initiating vascularization in gliomas. The combination of antiangiogenic agents with myeloid cell-targeting strategies might provide a new therapeutic approach for glioblastoma patients.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Myeloid Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Cell Line, Tumor , Coculture Techniques , Glioma/blood supply , Glioma/diagnosis , Glioma/pathology , Macrophages/metabolism , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Neovascularization, Pathologic/pathology
2.
J Neurooncol ; 121(2): 269-78, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25370707

ABSTRACT

Placenta growth factor (PlGF) is a member of vascular endothelial growth factor family which can promote cancer growth by various mechanisms. Placenta growth factor is upregulated in many neoplastic diseases and serum levels of PlGF are increased in cancer patients following anti-angiogenic therapy. However, its role in glioma growth is yet not fully elucidated. In this study we analyzed the expression of PlGF mRNA using real time PCR in human gliomas of different WHO grades. Placenta growth factor mRNA levels were highly variable and did not correlate with WHO grades, arguing against a significant role in glioma progression. The highest PlGF expression was observed in anaplastic astrocytomas whereas grade II astrocytomas and glioblastomas displayed lower levels of expression. Immunohistochemical analysis showed that PlGF was expressed by inflammatory and endothelial cells in addition to tumor cells. Placenta growth factor mRNA expression in 12 matched glioblastoma samples before and after therapy, including bevacizumab and cilengitide treatment was largely unaffected by the aforementioned treatment modalities. In vitro, the exposure of VEGFR-1 expressing glioma cells to bevacizumab did not increase the expression levels of PlGF mRNA. In summary, our results do not support the hypothesis that PlGF plays a major role in the resistance of gliomas after anti-angiogenic therapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm/physiology , Glioma/drug therapy , Pregnancy Proteins/metabolism , Adult , Aged , Antibodies, Monoclonal, Humanized/therapeutic use , Bevacizumab , Biomarkers, Tumor/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain/surgery , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Cell Line, Tumor , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Glioma/metabolism , Glioma/pathology , Glioma/surgery , Humans , Middle Aged , Neoplasm Grading , Placenta Growth Factor , Prognosis , RNA, Messenger/metabolism , Retrospective Studies , Snake Venoms/therapeutic use , Vascular Endothelial Growth Factor Receptor-1/metabolism
3.
Methods Mol Biol ; 1135: 275-88, 2014.
Article in English | MEDLINE | ID: mdl-24510872

ABSTRACT

The generation of bone marrow chimera in mice is a valuable tool to study a variety of cellular processes. Donor bone marrow cells expressing reporter genes have been used to study the process of cell differentiation and the mechanisms involved in bone marrow cell recruitment. Bone marrow cells bearing genetic manipulation have been used in bone marrow chimeras to elucidate the role of molecules in different physiological and pathological settings. Since in the normal adult brain angiogenesis does not occur, models of brain injury like ischemia and tumor growth have been used to study the contribution of bone marrow-derived cells to the cerebral vasculature. This chapter describes the procedures to perform bone marrow transplantation in order to study the contribution of bone marrow-derived cells to vascularization in an orthotopic glioma model.


Subject(s)
Hematopoietic Stem Cells/physiology , Neovascularization, Pathologic/pathology , Animals , Bone Marrow Cells/physiology , Bone Marrow Transplantation , Brain Neoplasms/blood supply , Cerebrovascular Circulation , Chimera , Glioma/blood supply , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation
4.
Int J Cancer ; 125(8): 1771-7, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19582874

ABSTRACT

Despite extensive research, the contribution of bone-marrow-derived endothelial progenitor cells (BM-EPC) to tumor angiogenesis remains controversial. In previous publications, the extent of incorporation of BM-EPCs into the endothelial cell (EC) layer in different tumor models has been reported as significant in some studies but undetectable in others. Here, we studied the differentiation of BM-EPCs and its contribution to tumor vessels in experimental and spontaneous lung metastasis (B16 melanoma and prostate carcinoma), in an autochthonous transgenic model of prostate tumorigenesis, in orthotopically implanted lung tumors [Lewis lung carcinoma (LLC)], in heterotopic subcutaneous models (LLC and C1 prostate carcinoma) growing in green fluorescent protein (GFP)-expressing bone marrow (BM) chimeras. Immunofluorescence was performed with a set of endothelial and hematopoietic markers and confocal microscopy was used to generate 3D reconstruction images. By performing rigorously conducted morphological studies, we found no evidence of BM-EPCs differentiation into tumor endothelium independently of tumor type, grade and organ site in primary and metastatic tumors. The vast majority of GFP(+) cells were trafficking leucocytes or periendothelial myeloid cells. To explore the possibility that local overexpression of vascular endothelial growth factor (VEGF) might increase the numbers of incorporated BM-EPCs, we analyzed tumors genetically manipulated to overexpress VEGF(164). Local VEGF production induces a massive infiltration of bone-marrow-derived cells, but did not lead to vessel wall integration of these cells. Collectively, these findings suggest that during tumor progression vascularization occurs primarily via classical tumor angiogenesis (e.g., sprouting of pre-existing ECs), whereas BM-EPCs do not incorporate into the vessel wall to any significant extent.


Subject(s)
Carcinoma, Lewis Lung/pathology , Endothelium, Vascular/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Prostatic Neoplasms/pathology , Stem Cells/pathology , Animals , Bone Marrow/pathology , Bone Marrow Transplantation , Humans , Immunoenzyme Techniques , Lung Neoplasms/therapy , Male , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Prostatic Neoplasms/therapy , Vascular Endothelial Growth Factor A/metabolism
5.
Cancer Res ; 68(18): 7342-51, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18794121

ABSTRACT

Several lines of evidence indicate that Flt-1, a fms-like tyrosine kinase receptor, which binds to vascular endothelial growth factor (VEGF)-A, VEGF-B, and PlGF, is a positive regulator of angiogenesis in the context of tumor growth and metastasis. However, the molecular basis of its action is still not clear. Besides endothelial cells, Flt-1 is also expressed by other different cell types, including myeloid hematopoeitic cells (monocytes and macrophages). To examine the functions of Flt-1 expressed by bone marrow-derived myeloid cells in supporting tumor growth and angiogenesis, Flt-1 tyrosine kinase-deficient (Flt-1 TK-/-) bone marrow cells were transplanted into lethally irradiated syngeneic recipients. After hematopoietic reconstitution, we orthotopically implanted syngeneic wild-type glioma cells or glioma cells overexpressing either VEGF(164) or PlGF-2. Loss of Flt-1 signaling in bone marrow-derived myeloid cells led to a significant decrease in tumor volume and vascularization in gliomas. VEGF but not PlGF overexpressed by glioma cells restored the tumor growth rate in Flt-1 TK-/- bone marrow chimera. VEGF and PlGF overexpression by tumor cells induced an accumulation of bone marrow-derived myeloid cells into tumor tissue. This infiltration was decreased in tumors grown in Flt-1 TK-/- bone marrow chimeras. When investigating chemokines and growth factors involved in myeloid cell recruitment, we determined elevated SDF-1/CXCL12 levels in VEGF- and PlGF-overexpressing tumors. Collectively, these results suggest that Flt-1 signaling in myeloid cells is essential to amplify the angiogenic response and to promote glioma growth.


Subject(s)
Glioma/enzymology , Glioma/pathology , Macrophages/enzymology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Bone Marrow Cells/enzymology , Bone Marrow Cells/pathology , Capillary Permeability/physiology , Cell Growth Processes/physiology , Chemokine CXCL12/biosynthesis , Chimera/metabolism , Glioma/blood supply , Glioma/metabolism , Macrophages/pathology , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/pathology , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Proteins/genetics , Proteins/metabolism , Signal Transduction , Transfection , Vascular Endothelial Growth Factor A/biosynthesis
6.
Neurosurgery ; 57(6): 1264-71; discussion 1264-71, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16331175

ABSTRACT

OBJECTIVE: Malignant gliomas are prominent targets for cancer gene therapy approaches because of their poor prognosis despite all available therapies. Endothelial cells (ECs) are considered attractive vehicles for cell-based gene therapy because of their tropism to the tumor vasculature. In this study, we investigated the potential of ECs to incorporate into glioma vessels after intra-arterial or local application to establish whether ECs can be used as cellular vectors for gene therapy in gliomas. METHODS: Immortalized rat brain endothelial cells (BECs) were modified to express either beta-galactosidase or green fluorescent protein (GFP). The ability of transduced BECs to integrate into tumor vessels after interstitial implantation was evaluated in C6 and 9L glioma models. The fate of GFP-BECs was investigated after selective intracarotid injection into C6 tumor-bearing animals. RESULTS: The interstitially grafted BECs organized themselves into vascular-like structures and integrated into the tumor vasculature. Transgene expression was limited to 10 days after injection. After selective intra-arterial injection, numerous GFP-BECs were adherent to the vascular lumen at least 7 days after injection. These cells were evenly distributed within small vessels and capillaries of the injected hemisphere and did not home selectively to the tumor vessels. CONCLUSION: Cell-based therapy approaches to brain tumor treatment using BECs as cellular vectors might be hampered by the rapid downregulation of transgene expression and by the fact that these cells do not home specifically to tumor vessels after intra-arterial injection. Nevertheless, locoregional administration of BECs might be an interesting approach for delivering molecules to brain tumors when short-term expression of transgene in the perivascular space is desirable.


Subject(s)
Brain Neoplasms/therapy , Brain/blood supply , Endothelial Cells/transplantation , Genetic Therapy/methods , Glioma/therapy , Animals , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation , Cell Survival , Endothelial Cells/metabolism , Endothelial Cells/physiology , Feasibility Studies , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Injections, Intra-Arterial , Luminescent Agents/metabolism , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Transduction, Genetic , Transfection , Transgenes
7.
Am J Pathol ; 165(5): 1557-70, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15509526

ABSTRACT

Angiopoietins have been implicated in playing an important role in blood vessel formation, remodeling, maturation, and maintenance. However, the role of angiopoietins in tumor angiogenesis remains uncertain. In this study, expression of human angiopoietin-1 (hAng-1) and angiopoietin (hAng-2) was amplified in the rat glioma cell line GS9L by stable transfection using an inducible tet-off system. Transfected cells were implanted intracerebrally into syngenic Fischer 344 rats. We demonstrated by means of magnetic resonance imaging that increased hAng-1 expression promoted a significant in vivo growth of intracerebral gliomas in rats. Overexpression of hAng-1 resulted in more numerous, more highly branched vessels, which were covered by pericytes. On the other hand, tumors derived from hAng-2-overexpressing cells were smaller than empty-plasmid control tumors. The tumor vasculature in these tumors was composed of aberrant small vascular cords, which were associated with few mural cells. Our results indicate that in the presence of hAng-1, tumors induce a more functional vascular network, which led to better tumor perfusion and growth. On the other hand, overexpression of hAng-2 led to less intact tumor vessels, inhibited capillary sprouting, and impaired tumor growth.


Subject(s)
Angiopoietin-1/physiology , Brain Neoplasms/pathology , Glioma/pathology , Neovascularization, Pathologic , Animals , Apoptosis , Blotting, Northern , Blotting, Western , Brain/pathology , Capillaries/pathology , Cell Line, Tumor , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Evans Blue/pharmacology , Fluorescent Antibody Technique, Indirect , Genetic Vectors , In Situ Hybridization , In Situ Nick-End Labeling , Magnetic Resonance Imaging , Microscopy, Confocal , Microscopy, Fluorescence , Plasmids/metabolism , Rats , Rats, Inbred F344 , Software
9.
Brain Pathol ; 13(4): 582-97, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14655762

ABSTRACT

Until recently, it was generally accepted that the vascularization of solid tumors occurred exclusively through the sprouting and co-option from pre-existing blood vessels. Growing evidence now suggests that bone marrow-derived endothelial progenitor cells (EP) circulate in the blood and may play an important role in the formation of new blood vessels in certain tumors. Whether endothelial progenitors participate in the vascularization of brain tumors has not yet been evaluated. In this study, we examined the contribution of EP to tumor angiogenesis in a murine glioma tumor model. Donor bone marrow cells obtained from transgenic mice constitutively expressing beta-galactosidase or GFP either ubiquitously or transcriptionally regulated by an endothelial specific promotor Tie-2 were injected into lethally irradiated adult mice. After bone marrow reconstitution by donor cells, mice were implanted with syngeneic GL261 murine glioma cells. Morphological and confocal 3-dimensional analysis showed that the majority of the engrafted donor marrow cells were expressing hematopoietic and/or microglia markers, but did not appreciably contribute to the tumor vasculature. Implantation of glioma cells genetically engineered to overexpress VEGF produced highly vascularized tumors. However, the number of endothelial progenitors incorporated in the tumor vasculature did not increase. These data strongly suggest that neovascularization in the brain might fundamentally be regulated by the sprouting of pre-existing vessels and implicate that circulating endothelial progenitors do not play a significant role in this process.


Subject(s)
Glioma/physiopathology , Hematopoietic Stem Cells/physiology , Neovascularization, Pathologic/physiopathology , Animals , Antigens, CD , Antigens, Differentiation/metabolism , Blotting, Northern , Bone Marrow/physiology , CD11b Antigen/metabolism , Cell Line, Tumor , Endoglin , Glial Fibrillary Acidic Protein/metabolism , Glioma/metabolism , Glioma/veterinary , Glycoside Hydrolases/metabolism , Hematopoietic Stem Cell Transplantation/methods , Immunohistochemistry , Infections , Lac Operon , Leukocyte Common Antigens/metabolism , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Microscopy, Confocal , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA, Messenger/biosynthesis , Receptors, Cell Surface , Reverse Transcriptase Polymerase Chain Reaction/methods , Spleen/metabolism , Time Factors , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , von Willebrand Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...