Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Opt Lett ; 38(15): 2786-8, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23903142

ABSTRACT

We report an accurate, precise and sensitive method and system for quantitative fluorescence image-guided neurosurgery. With a low-noise, high-dynamic-range CMOS array, we perform rapid (integration times as low as 50 ms per wavelength) hyperspectral fluorescence and diffuse reflectance detection and apply a correction algorithm to compensate for the distorting effects of tissue absorption and scattering. Using this approach, we generated quantitative wide-field images of fluorescence in tissue-simulating phantoms for the fluorophore PpIX, having concentrations and optical absorption and scattering variations over clinically relevant ranges. The imaging system was tested in a rodent model of glioma, detecting quantitative levels down to 20 ng/ml. The resulting performance is a significant advance on existing wide-field quantitative imaging techniques, and provides performance comparable to a point-spectroscopy probe that has previously demonstrated significant potential for improved detection of malignant brain tumors during surgical resection.


Subject(s)
Neurosurgery/methods , Surgery, Computer-Assisted/methods , Animals , Brain/metabolism , Brain/pathology , Glioma/pathology , Glioma/surgery , Protoporphyrins/metabolism , Rats , Spectrometry, Fluorescence
2.
J Neurooncol ; 114(1): 33-42, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23695515

ABSTRACT

Glioblastoma multiform is one of the most common and aggressive primary brain tumors in adults. High glutamate levels are thought to contribute to glioma growth. While research has focused on understanding glutamate signaling in glioma cells, little is known about the role of glutamate between glioma and astrocyte interactions. To study the relationship between astrocytes and tumor cells, the CNS-1 rodent glioma cell line was used. We hypothesized increased glutamate uptake by astrocytes would negatively affect CNS-1 cell growth. Primary rodent astrocytes and CNS-1 cells were co-cultured for 7 days in a Boyden chamber in the presence of 5 mM glutamate. Cells were treated with propentofylline, an atypical synthetic methylxanthine known to increase glutamate transporter expression in astrocytes. Our results indicate astrocytes can increase glutamate uptake through the GLT-1 transporter, leading to less glutamate available for CNS-1 cells, ultimately resulting in increased CNS-1 cell apoptosis. These data suggest that astrocytes in the tumor microenvironment can be targeted by the drug, propentofylline, affecting tumor cell growth.


Subject(s)
Apoptosis/drug effects , Astrocytes/metabolism , Glutamic Acid/metabolism , Neuroprotective Agents/pharmacology , Xanthines/pharmacology , Animals , Animals, Newborn , Annexin A5/metabolism , Astrocytes/drug effects , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Cerebral Cortex/cytology , Coculture Techniques , Dose-Response Relationship, Drug , Excitatory Amino Acid Transporter 2 , Gene Expression Regulation, Neoplastic/drug effects , Glioma/pathology , Glutamate Plasma Membrane Transport Proteins/genetics , Glutamic Acid/pharmacology , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Inbred Lew
3.
Sci Rep ; 2: 798, 2012.
Article in English | MEDLINE | ID: mdl-23152935

ABSTRACT

Intraoperative visual fluorescence imaging (vFI) has emerged as a promising aid to surgical guidance, but does not fully exploit the potential of the fluorescent agents that are currently available. Here, we introduce a quantitative fluorescence imaging (qFI) approach that converts spectrally-resolved data into images of absolute fluorophore concentration pixel-by-pixel across the surgical field of view (FOV). The resulting estimates are linear, accurate, and precise relative to true values, and spectral decomposition of multiple fluorophores is also achieved. Experiments with protoporphyrin IX in a glioma rodent model demonstrate in vivo quantitative and spectrally-resolved fluorescence imaging of infiltrating tumor margins for the first time. Moreover, we present images from human surgery which detect residual tumor not evident with state-of-the-art vFI. The wide-field qFI technique has broad implications for intraoperative surgical guidance because it provides near real-time quantitative assessment of multiple fluorescent biomarkers across the operative field.


Subject(s)
Brain Neoplasms/surgery , Fluorescence , Optical Imaging/methods , Animals , Brain Neoplasms/diagnosis , Fluorescent Dyes , Glioma/diagnosis , Glioma/surgery , Humans , Intraoperative Period , Optical Imaging/instrumentation , Phantoms, Imaging , Protoporphyrins , Rats
4.
PLoS One ; 7(5): e37955, 2012.
Article in English | MEDLINE | ID: mdl-22649568

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and aggressive primary brain cancer, with a median survival of less than 2 years after diagnosis with current available therapies. The tumor microenvironment serves a critical role in tumor invasion and progression, with microglia as a critical player. Our laboratory has previously demonstrated that propentofylline, an atypical methylxanthine with central nervous system glial modulating and anti-inflammatory actions, significantly decreases tumor growth in a GBM rodent model by preferentially targeting microglia. In the present study, we used the CNS-1 rat glioma model to elucidate the mechanisms of propentofylline. Here we demonstrate that propentofylline targets TROY, a novel signaling molecule up-regulated in infiltrating microglia, and not macrophages, in response to CNS-1 cells. We identify Pyk2, Rac1 and pJNK as the downstream signaling molecules of TROY through western blot analysis and siRNA transfection. We demonstrate that inhibition of TROY expression in microglia by siRNA transfection significantly inhibits microglial migration towards CNS-1 cells similar to 10 µM propentofylline treatment. These results identify TROY as a novel molecule expressed in microglia, involved in their migration and targeted by propentofylline. Furthermore, these results describe a signaling molecule that is differentially expressed between microglia and macrophages in the tumor microenvironment.


Subject(s)
Glioblastoma/physiopathology , Microglia/physiology , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/physiology , Xanthines/pharmacology , Animals , Blotting, Western , Cell Movement/drug effects , Cell Movement/physiology , Flow Cytometry , Focal Adhesion Kinase 2/metabolism , Glioblastoma/drug therapy , Immunohistochemistry , Microglia/drug effects , Microglia/metabolism , Neuropeptides/metabolism , RNA Interference , RNA, Small Interfering/genetics , Rats , Signal Transduction/genetics , Xanthines/therapeutic use , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
5.
Neuro Oncol ; 14(2): 119-31, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22086978

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and aggressive primary brain cancer, with a median survival of less than 2 years after diagnosis. The tumor microenvironment plays a critical role in tumor invasion and progression. Microglia and infiltrating macrophages are the most abundant immune cells in the tumor. In the present study, we demonstrate that systemic propentofylline (PPF), an atypical methylxanthine with central nervous system (CNS) glial modulating and anti-inflammatory actions, significantly decreased tumor growth in a CNS-1 rat model of GBM by targeting microglia and not tumor cells. Rats received tumor injections of 1 × 10(5) CNS-1 cells in the right striatum with daily intraperitonial injections of PPF (50 mg/kg) or saline beginning the day of tumor injection. PPF did not cause apoptosis or decrease proliferation of CNS-1 tumor cells. Furthermore, we demonstrate, using in vitro methods, that PPF decreased microglial migration toward CNS-1 tumor cells and decreased MMP-9 expression. The effects of PPF were shown to be specific to microglia and not peripheral macrophages. These results support a differential functional role of resident microglia and infiltrating macrophages in the brain tumor environment. Our data highlight microglia as a crucial target for future therapeutic development and present PPF as a possible drug for treatment of human GBM.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Macrophages/drug effects , Microglia/drug effects , Xanthines/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Central Nervous System/drug effects , Central Nervous System/immunology , Central Nervous System/metabolism , Disease Models, Animal , Glioblastoma/immunology , Glioblastoma/metabolism , Humans , Macrophages/immunology , Male , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/metabolism , Microglia/pathology , Neoplasm Invasiveness/pathology , Rats , Tumor Cells, Cultured , Tumor Microenvironment , Xanthines/therapeutic use
6.
Exp Neurol ; 234(2): 340-50, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22119425

ABSTRACT

There is a growing body of preclinical evidence for the potential involvement of glial cells in neuropathic pain conditions. Several glial-targeted agents are in development for the treatment of pain conditions. Here we report the failure of a glial modulating agent, propentofylline, to decrease pain reported in association with post-herpetic neuralgia. We offer new evidence to help explain why propentofylline failed in patients by describing in vitro functional differences between rodent and human microglia and macrophages. We directly compared the proinflammatory response induced by lipopolysaccharide (LPS) with or without propentofylline using rat postnatal microglia, rat peritoneal macrophages, human fetal microglia, human peripheral macrophages and human immortalized THP-1 cells. We measured tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß) and nitrite release (as an indicator of nitric oxide (NO)) as downstream indicators. We found that LPS treatment did not induce nitrite in human microglia, macrophages or THP-1 cells; however LPS treatment did induce nitrite release in rat microglia and macrophages. Following LPS exposure, propentofylline blocked TNF-α release in rodent microglia with all the doses tested (1-100 µM), and dose-dependently decreased TNF-α release in rodent macrophages. Propentofylline partially decreased TNF-α (35%) at 100 µM in human microglia, macrophages and THP-1 macrophages. Propentofylline blocked nitrite release from LPS stimulated rat microglia and inhibited nitrite in LPS-stimulated rat macrophages. IL-1ß was decreased in LPS-stimulated human microglia following propentofylline at 100 µM. Overall, human microglia were less responsive to LPS stimulation and propentofylline treatment than the other cell types. Our data demonstrate significant functional differences between cell types and species following propentofylline treatment and LPS stimulation. These results may help explain the differential behavioral effects of propentofylline observed between rodent models of pain and the human clinical trial.


Subject(s)
Macrophages/drug effects , Microglia/drug effects , Neuralgia, Postherpetic/drug therapy , Xanthines/pharmacology , Animals , Humans , Macrophages/metabolism , Microglia/metabolism , Neuralgia, Postherpetic/immunology , Neuralgia, Postherpetic/physiopathology , Tumor Necrosis Factor-alpha/metabolism , Xanthines/therapeutic use
7.
ASN Neuro ; 3(3): e00063, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21740400

ABSTRACT

GBM (glioblastoma multiforme) is a highly aggressive brain tumour with very poor prognosis despite multi-modalities of treatment. Furthermore, recent failure of targeted therapy for these tumours highlights the need of appropriate rodent models for preclinical studies. In this review, we highlight the most commonly used rodent models (U251, U86, GL261, C6, 9L and CNS-1) with a focus on the pathological and genetic similarities to the human disease. We end with a comprehensive review of the CNS-1 rodent model.


Subject(s)
Brain Neoplasms/pathology , Disease Models, Animal , Glioblastoma/pathology , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Clinical Trials as Topic , Glioblastoma/genetics , Humans
8.
J Immunol ; 186(1): 275-83, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21106849

ABSTRACT

Treatment of tumor-bearing mice with a stimulatory Ab to glucocorticoid-induced TNFR family-related receptor (GITR) has previously been shown to elicit protective T cell responses against poorly immunogenic tumors. However, the role of GITR stimulation on CD8 T cells and the nature of tumor rejection Ags have yet to be determined. In this study, we show that a stimulatory mAb to GITR (clone DTA-1) acts directly on CD8 T cells, but not on CD4(+)CD25(+) regulatory T (T(reg)) cells, in B16 tumor-bearing mice to induce concomitant immunity against secondary B16 tumors, as well as protective memory following surgical excision of the primary tumor. Melanoma growth itself induced GITR expression on tumor-specific CD8 T cells, providing a mechanism whereby these cells may respond to stimulatory anti-GITR. Unexpectedly, in contrast to T(reg) cell depletion therapy with anti-CD4, GITR stimulation induced very weak CD8 T cell responses to melanocyte differentiation Ags expressed by the tumor, and did not induce autoimmune vitiligo. Accordingly, GITR-stimulated hosts that were primed with B16 melanoma rejected B16, but not the unrelated JBRH melanoma, indicating that tumor rejection Ags are tumor-specific rather than shared. In support of this, we show that GITR stimulation induces CD8 T cell responses to a tumor-specific Ag, and that these responses are of higher functional avidity compared with those induced by T(reg) cell depletion. We conclude that stimulation of GITR on effector CD8 T cells results in high-avidity T cell responses to tumor-specific Ags, thereby inducing potent antitumor immunity in the absence of autoimmunity.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/immunology , Lymphocyte Activation/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Receptors, Nerve Growth Factor/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Adhesion/immunology , Cell Line, Tumor , Female , Glucocorticoid-Induced TNFR-Related Protein , Lymphocyte Depletion , Male , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/immunology , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...