Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Neurooncol ; 99(1): 13-24, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20063114

ABSTRACT

Malignant primary glial and secondary metastatic brain tumors represent distinct pathological entities. Nevertheless, both tumor types induce profound angiogenic responses in the host brain microvasculature that promote tumor growth. We hypothesized that primary and metastatic tumors induce similar microvascular changes that could function as conserved angiogenesis based therapeutic targets. We previously isolated glioma endothelial marker genes (GEMs) that were selectively upregulated in the microvasculature of proliferating glioblastomas. We sought to determine whether these genes were similarly induced in the microvasculature of metastatic brain tumors. RT-PCR and quantitative RT-PCR were used to screen expression levels of 20 candidate GEMs in primary and metastatic clinical brain tumor specimens. Differentially regulated GEMs were further evaluated by immunohistochemistry or in situ hybridization to localize gene expression using clinical tissue microarrays. Thirteen GEMs were upregulated to a similar degree in both primary and metastatic brain tumors. Most of these genes localize to the cell surface (CXCR7, PV1) or extracellular matrix (COL1A1, COL3A1, COL4A1, COL6A2, MMP14, PXDN) and were selectively expressed by the microvasculature. The shared expression profile between primary and metastatic brain tumors suggests that the molecular pathways driving the angiogenic response are conserved, despite differences in the tumor cells themselves. Anti-angiogenic therapies currently in development for primary brain tumors may prove beneficial for brain metastases and vice versa.


Subject(s)
Brain Neoplasms/metabolism , Collagen/metabolism , Gene Expression Regulation, Neoplastic/physiology , Glioma/metabolism , Matrix Metalloproteinase 14/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chi-Square Distribution , Collagen/classification , Collagen/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Glioma/genetics , Glioma/pathology , Glioma/secondary , Humans , Matrix Metalloproteinase 14/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Peroxidase/genetics , Peroxidase/metabolism , RNA, Messenger/metabolism , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Peroxidasin
2.
BMC Cancer ; 9: 417, 2009 Nov 30.
Article in English | MEDLINE | ID: mdl-19948061

ABSTRACT

BACKGROUND: TEM1/endosialin is an emerging microvascular marker of tumor angiogenesis. We characterized the expression pattern of TEM1/endosialin in astrocytic and metastatic brain tumors and investigated its role as a therapeutic target in human endothelial cells and mouse xenograft models. METHODS: In situ hybridization (ISH), immunohistochemistry (IH) and immunofluorescence (IF) were used to localize TEM1/endosialin expression in grade II-IV astrocytomas and metastatic brain tumors on tissue microarrays. Changes in TEM1/endosialin expression in response to pro-angiogenic conditions were assessed in human endothelial cells grown in vitro. Intracranial U87MG glioblastoma (GBM) xenografts were analyzed in nude TEM1/endosialin knockout (KO) and wildtype (WT) mice. RESULTS: TEM1/endosialin was upregulated in primary and metastatic human brain tumors, where it localized primarily to the tumor vasculature and a subset of tumor stromal cells. Analysis of 275 arrayed grade II-IV astrocytomas demonstrated TEM1/endosialin expression in 79% of tumors. Robust TEM1/endosialin expression occurred in 31% of glioblastomas (grade IV astroctyomas). TEM1/endosialin expression was inversely correlated with patient age. TEM1/endosialin showed limited co-localization with CD31, alphaSMA and fibronectin in clinical specimens. In vitro, TEM1/endosialin was upregulated in human endothelial cells cultured in matrigel. Vascular Tem1/endosialin was induced in intracranial U87MG GBM xenografts grown in mice. Tem1/endosialin KO vs WT mice demonstrated equivalent survival and tumor growth when implanted with intracranial GBM xenografts, although Tem1/endosialin KO tumors were significantly more vascular than the WT counterparts. CONCLUSION: TEM1/endosialin was induced in the vasculature of high-grade brain tumors where its expression was inversely correlated with patient age. Although lack of TEM1/endosialin did not suppress growth of intracranial GBM xenografts, it did increase tumor vascularity. The cellular localization of TEM1/endosialin and its expression profile in primary and metastatic brain tumors support efforts to therapeutically target this protein, potentially via antibody mediated drug delivery strategies.


Subject(s)
Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Age Factors , Animals , Brain Neoplasms/blood supply , Fluorescent Antibody Technique , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Mice, Nude , Neovascularization, Pathologic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Xenograft Model Antitumor Assays
3.
Cancer Res ; 69(12): 4999-5006, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19491259

ABSTRACT

Defective apoptosis contributes to tumorigenesis, although the critical molecular targets remain to be fully characterized. PUMA, a BH3-only protein essential for p53-dependent apoptosis, has been shown to suppress lymphomagenesis. In this study, we investigated the role of PUMA in intestinal tumorigenesis using two animal models. In the azoxymethane (AOM)/dextran sulfate sodium salt model, PUMA deficiency increased the multiplicity and size of colon tumors but reduced the frequency of beta-catenin hotspot mutations. The absence of PUMA led to a significantly elevated incidence of precursor lesions induced by AOM. AOM was found to induce p53-dependent PUMA expression and PUMA-dependent apoptosis in the colonic crypts and stem cell compartment. Furthermore, PUMA deficiency significantly enhanced the formation of spontaneous macroadenomas and microadenomas in the distal small intestine and colon of APC(Min/+) mice. These results show an essential role of PUMA-mediated apoptosis in suppressing intestinal tumorigenesis in mice.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Intestinal Neoplasms/pathology , Proto-Oncogene Proteins/physiology , Animals , Apoptosis , Blotting, Western , Genes, APC , In Situ Hybridization , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , beta Catenin/genetics
4.
Cell Stem Cell ; 2(6): 576-83, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18522850

ABSTRACT

Radiation is one of the most effective cancer treatments. However, gastrointestinal (GI) syndrome is a major limiting factor in abdominal and pelvic radiotherapy. The loss of crypt stem cells or villus endothelial cells has been suggested to be responsible for radiation-induced intestinal damage. We report here a critical role of the BH3-only protein p53 upregulated modulator of apoptosis (PUMA) in the radiosensitivity of intestinal epithelium and pathogenesis of GI syndrome. PUMA was induced in a p53-dependent manner and mediated radiation-induced apoptosis via the mitochondrial pathway in the intestinal mucosa. PUMA-deficient mice exhibited blocked apoptosis in the intestinal progenitor and stem cells, enhanced crypt proliferation and regeneration, and prolonged survival following lethal doses of radiation. Unexpectedly, PUMA deficiency had little effect on radiation-induced intestinal endothelial apoptosis. Suppressing PUMA expression by antisense oligonucleotides provided significant intestinal radioprotection. Therefore, PUMA-mediated apoptosis in the progenitor and stem cell compartments is crucial for radiation-induced intestinal damage.


Subject(s)
Adult Stem Cells/radiation effects , Apoptosis Regulatory Proteins/genetics , Gastrointestinal Tract/radiation effects , Radiation Injuries, Experimental/genetics , Tumor Suppressor Proteins/genetics , Adult Stem Cells/cytology , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/biosynthesis , Gastrointestinal Tract/cytology , Gene Expression Regulation/genetics , Gene Expression Regulation/radiation effects , Intestinal Mucosa/cytology , Intestinal Mucosa/radiation effects , Mice , Neoplasms/radiotherapy , Oligonucleotides, Antisense , Radiation Injuries, Experimental/pathology , Signal Transduction/radiation effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/radiation effects , Tumor Suppressor Proteins/biosynthesis , Whole-Body Irradiation
5.
BMC Neurosci ; 9: 29, 2008 Feb 26.
Article in English | MEDLINE | ID: mdl-18302779

ABSTRACT

BACKGROUND: Plasmalemmal vesicle associated protein-1 (PV-1) is selectively expressed in human brain microvascular endothelial cells derived from clinical specimens of primary and secondary malignant brain tumors, cerebral ischemia, and other central nervous system (CNS) diseases associated with blood-brain barrier breakdown. In this study, we characterize the murine CNS expression pattern of PV-1 to determine whether localized PV-1 induction is conserved across species and disease state. RESULTS: We demonstrate that PV-1 is selectively upregulated in mouse blood vessels recruited by brain tumor xenografts at the RNA and protein levels, but is not detected in non-neoplastic brain. Additionally, PV-1 is induced in a mouse model of acute ischemia. Expression is confined to the cerebovasculature within the region of infarct and is temporally regulated. CONCLUSION: Our results confirm that PV-1 is preferentially induced in the endothelium of mouse brain tumors and acute ischemic brain tissue and corresponds to blood-brain barrier disruption in a fashion analogous to human patients. Characterization of PV-1 expression in mouse brain is the first step towards development of rodent models for testing anti-edema and anti-angiogenesis therapeutic strategies based on this molecule.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Ischemia/metabolism , Brain Neoplasms/metabolism , Carrier Proteins/analysis , Endothelium, Vascular/metabolism , Membrane Proteins/analysis , Animals , Biomarkers/analysis , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Up-Regulation
6.
J Neurotrauma ; 24(8): 1347-54, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17711396

ABSTRACT

Serum magnesium concentration has a neuroprotective effect in experimental models of traumatic brain injury (TBI). This study was designed to assess the relationship between initial serum magnesium, cerebrospinal fluid (CSF) magnesium, neurological outcome and the efficacy of magnesium replacement therapy (MgSO4). A retrospective analysis was performed on a prospectively collected dataset from 216 patients admitted during 1996-2006 to the University of Pittsburgh Medical Center with severe TBI. Admission serum and CSF magnesium were dichotomized into low and normal magnesium concentration groups for serum and normal and high concentration groups for CSF. A logistic-regression analysis was performed with 6-month Glasgow Outcome Scale (GOS) scores as outcome variable. The outcome of a subset of 31 patients who presented with low serum magnesium and who were rapidly corrected within 24 h of admission was also analyzed. Low initial serum magnesium was measured in 56.67% of all patients. Patients with an initial serum magnesium of <1.3 mEq/L were 2.37 times more likely to have a poor outcome (CI: 1.18-4.78, p = 0.016). The prognostic significance of depressed serum magnesium remained, even in patients whose serum magnesium levels were corrected within 24 h (OR = 11.03, CI: 1.87-68.14, p = 0.008). Patients with an initial high CSF magnesium were 7.63 more likely to have a poor outcome (p = 0.05). Elevated CSF magnesium correlated with depressed serum magnesium only in patients with poor outcome (p = 0.013). Patients with low serum magnesium and high CSF magnesium are most likely to have poor outcome after severe TBI. Rapid correction of serum magnesium levels does not reverse the prognostic value of these markers.


Subject(s)
Anticonvulsants/therapeutic use , Brain Injuries/drug therapy , Brain Injuries/metabolism , Magnesium Sulfate/therapeutic use , Magnesium/blood , Magnesium/cerebrospinal fluid , Adolescent , Adult , Aged , Cohort Studies , Databases, Factual , Female , Glasgow Coma Scale , Humans , Male , Middle Aged , Retrospective Studies , Treatment Outcome
7.
Clin Cancer Res ; 11(21): 7643-50, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16278383

ABSTRACT

PURPOSE: Plasmalemmal vesicle associated protein-1 (PV-1) is up-regulated in the endothelium of human glioblastoma. We sought to further characterize the expression pattern of PV-1 in human brain tumors and interrogate its role in brain tumor angiogenesis. EXPERIMENTAL DESIGN: Quantitative reverse transcription-PCR and in situ hybridization were used to measure PV-1 expression in a panel of 46 human brain tumors and related pathologic states. Matrigel tubulogenesis assays and cell migration assays were used to show function of PV-1 in primary human endothelial cells (HMVEC) under gene knockdown conditions. RESULTS: PV-1 is selectively up-regulated in a variety of high-grade human brain tumors, including glioblastoma and metastatic carcinoma, as well as other cerebral disorders associated with blood-brain barrier disruption, such as acute ischemia. Expression levels were reduced in low-grade neoplasia; however, tumors associated with the ependyma and choroid plexus, known sites of PV-1 expression, also exhibited robust expression. Cerebral expression of PV-1 mRNA was confined to endothelial cells in all cases. PV-1 expression was induced in HMVEC cells in vitro by exposure to medium conditioned by U87MG and U251MG human brain tumor cell lines and by medium supplemented with exogenous vascular endothelial growth factor or scatter factor/hepatocyte growth factor. RNA interference-mediated inhibition of PV-1 induction in HMVEC cells blocked Matrigel-induced tubulogenesis and inhibited cell migration induced by conditioned medium or angiogenic growth factors. CONCLUSIONS: Our results confirm that PV-1 is preferentially induced in the endothelium of high-grade human brain tumors. Inhibition of PV-1 expression is associated with failure of endothelial differentiation in vitro. PV-1 represents a novel marker of brain tumor angiogenesis and integrity of the blood-brain barrier and is a potential therapeutic target.


Subject(s)
Biomarkers, Tumor , Brain Neoplasms/pathology , Carrier Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Membrane Proteins/biosynthesis , Neovascularization, Pathologic , Blood-Brain Barrier , Blotting, Western , Cell Line, Tumor , Cell Movement , Cells, Cultured , Collagen/chemistry , Culture Media, Conditioned/pharmacology , Drug Combinations , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Humans , In Situ Hybridization , Ischemia , Laminin/chemistry , Microcirculation , Neoplasm Metastasis , Proteoglycans/chemistry , RNA/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
8.
Am J Pathol ; 165(2): 601-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277233

ABSTRACT

Malignant gliomas are uniformly lethal tumors whose morbidity is mediated in large part by the angiogenic response of the brain to the invading tumor. This profound angiogenic response leads to aggressive tumor invasion and destruction of surrounding brain tissue as well as blood-brain barrier breakdown and life-threatening cerebral edema. To investigate the molecular mechanisms governing the proliferation of abnormal microvasculature in malignant brain tumor patients, we have undertaken a cell-specific transcriptome analysis from surgically harvested nonneoplastic and tumor-associated endothelial cells. SAGE-derived endothelial cell gene expression patterns from glioma and nonneoplastic brain tissue reveal distinct gene expression patterns and consistent up-regulation of certain glioma endothelial marker genes across patient samples. We define the G-protein-coupled receptor RDC1 as a tumor endothelial marker whose expression is distinctly induced in tumor endothelial cells of both brain and peripheral vasculature. Further, we demonstrate that the glioma-induced gene, PV1, shows expression both restricted to endothelial cells and coincident with endothelial cell tube formation. As PV1 provides a framework for endothelial cell caveolar diaphragms, this protein may serve to enhance glioma-induced disruption of the blood-brain barrier and transendothelial exchange. Additional characterization of this extensive brain endothelial cell gene expression database will provide unique molecular insights into vascular gene expression.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Endothelium, Vascular/metabolism , Glioma/metabolism , Neovascularization, Pathologic/genetics , Biomarkers, Tumor/genetics , Brain/blood supply , Brain Neoplasms/pathology , Endothelium, Vascular/pathology , Glioma/pathology , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Cancer Res ; 64(3): 817-20, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871805

ABSTRACT

Tumor endothelial marker (TEM)8 was uncovered as a gene expressed predominantly in tumor endothelium, and its protein product was recently identified as the receptor for anthrax toxin. Here, we demonstrate that TEM8 protein is preferentially expressed in endothelial cells of neoplastic tissue. We used the extracellular domain of TEM8 to search for ligands and identified the alpha 3 subunit of collagen VI as an interacting partner. The TEM8-interacting region on collagen alpha 3(VI) was mapped to its COOH-terminal C5 domain. Remarkably, collagen alpha 3(VI) is also preferentially expressed in tumor endothelium in a pattern concordant with that of TEM8. These results suggest that the TEM8/C5 interaction may play an important biological role in tumor angiogenesis.


Subject(s)
Collagen Type VI/metabolism , Receptors, Cell Surface/metabolism , Endothelium, Vascular/metabolism , Humans , Membrane Proteins , Microfilament Proteins , Neoplasm Proteins , Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Protein Structure, Tertiary , Receptors, Cell Surface/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL