Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Neurooncol ; 110(2): 163-77, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22875710

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and devastating form of primary central nervous system malignancy. The prognosis for patients diagnosed with GBM is poor, having a median survival rate of 12-15 months. Despite modern advances in the development of antineoplastic agents, the efficacy of newer anti-cancer agents in the treatment of GBM is yet to be determined. Thus, there remains a significant unmet need for new therapeutic strategies against GBM. A promising chemotherapeutic intervention has emerged from studies of cannabinoid receptor agonists wherein tetrahydrocannabinol has been the most extensively studied. The novel cannabinoid ligand KM-233 was developed as a lead platform for future optimization of biopharmaceutical properties of classical based cannabinoid ligands. Treatment of U87MG human GBM cells with KM-233 caused a time dependent change in the phosphorylation profiles of MEK, ERK1/2, Akt, BAD, STAT3, and p70S6K. Almost complete mitochondrial depolarization was observed 6 h post-treatment followed by a rapid increase in cleaved caspase 3 and significant cytoskeletal contractions. Treatment with KM-233 also resulted in a redistribution of the Golgi-endoplasmic reticulum structures. Dose escalation studies in the orthotopic model using U87MG cells revealed an 80 % reduction in tumor size after 12 mg/kg daily dosing for 20 days. The evaluation of KM-233 against primary tumor tissue in the side flank model revealed a significant decrease in the rate of tumor growth. These findings indicate that structural refinement of KM-233 to improve its biopharmaceutical properties may lead to a novel and efficacious treatment for GBM.


Subject(s)
Brain Neoplasms/drug therapy , Cannabinoids/therapeutic use , Glioma/drug therapy , Animals , Apoptosis/drug effects , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Caspase 3/metabolism , Disease Models, Animal , Female , Glioma/metabolism , Glioma/pathology , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Mice, SCID , Phosphoproteins/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Tumor Cells, Cultured
2.
Int J Radiat Oncol Biol Phys ; 82(3): 1075-82, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22197233

ABSTRACT

PURPOSE: Damage to normal tissue is a limiting factor in clinical radiotherapy (RT). We tested the hypothesis that the presence of tumor alters the response of normal tissues to irradiation using a rat in situ brain tumor model. METHODS AND MATERIALS: Intravital microscopy was used with a rat cranial window to assess the in situ effect of rat C6 glioma on peritumoral tissue with and without RT. The RT regimen included 40 Gy at 8 Gy/day starting Day 5 after tumor implant. Endpoints included blood-brain barrier permeability, clearance index, leukocyte-endothelial interactions and staining for vascular endothelial growth factor (VEGF) glial fibrillary acidic protein, and apoptosis. To characterize the system response to RT, animal survival and tumor surface area and volume were measured. Sham experiments were performed on similar animals implanted with basement membrane matrix absent of tumor cells. RESULTS: The presence of tumor alone increases permeability but has little effect on leukocyte-endothelial interactions and astrogliosis. Radiation alone increases tissue permeability, leukocyte-endothelial interactions, and astrogliosis. The highest levels of permeability and cell adhesion were seen in the model that combined tumor and irradiation; however, the presence of tumor appeared to reduce the volume of rolling leukocytes. Unirradiated tumor and peritumoral tissue had poor clearance. Irradiated tumor and peritumoral tissue had a similar clearance index to irradiated and unirradiated sham-implanted animals. Radiation reduces the presence of VEGF in peritumoral normal tissues but did not affect the amount of apoptosis in the normal tissue. Apoptosis was identified in the tumor tissue with and without radiation. CONCLUSIONS: We developed a novel approach to demonstrate that the presence of the tumor in a rat intracranial model alters the response of normal tissues to irradiation.


Subject(s)
Blood-Brain Barrier/radiation effects , Brain Neoplasms/radiotherapy , Brain/radiation effects , Endothelium, Vascular/radiation effects , Glioma/radiotherapy , Animals , Apoptosis , Blood-Brain Barrier/physiology , Brain/blood supply , Brain/metabolism , Brain Neoplasms/blood supply , Brain Neoplasms/chemistry , Brain Neoplasms/pathology , Cell Adhesion/radiation effects , Coloring Agents/pharmacokinetics , Dose Fractionation, Radiation , Endothelium, Vascular/physiology , Glioma/blood supply , Glioma/chemistry , Glioma/pathology , Gliosis/pathology , Leukocyte Rolling/physiology , Leukocyte Rolling/radiation effects , Leukocytes/physiology , Leukocytes/radiation effects , Male , Models, Animal , Permeability/radiation effects , Rats , Rats, Wistar , Tumor Burden/radiation effects , Vascular Endothelial Growth Factor A/analysis
3.
Surgery ; 150(3): 497-504, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21878236

ABSTRACT

BACKGROUND: High-grade glioblastomas have immature, leaky tumor blood vessels that impede the efficacy of adjuvant therapy. We assessed the ability of human interferon (hIFN)-ß delivered locally via gene transfer to effect vascular stabilization in an orthotopic model of glioblastoma xenograft resection. METHODS: Xenografts were established by injecting 3 grade IV glioblastoma cell lines (GBM6-luc, MT330-luc, and SJG2-luc) into the cerebral cortex of nude rats. Tumors underwent subtotal resection, and then had gel foam containing an adeno-associated virus vector encoding either hIFN-ß or green fluorescence protein (control) placed in the resection cavity. The primary endpoint was stabilization of tumor vasculature, as evidenced by CD34, α-SMA, and CA IX staining. Overall survival was a secondary endpoint. RESULTS: hIFN-ß treatment altered the tumor vasculature of GBM6-luc and SJG2-luc xenografts, decreasing the density of endothelial cells, stabilizing vessels with pericytes, and decreasing tumor hypoxia. The mean survival for rats with these neoplasms was not improved, however. In rats with MT330-luc xenografts, hIFN-ß resulted in tumor regression with a 6-month survival of 55% (INF-ß group) and 9% (control group). CONCLUSION: The use of AAV hIFN-ß in our orthotopic model of glioblastoma resection stabilized tumor vasculature and improved survival in rats with MT330 xenografts.


Subject(s)
Brain Neoplasms/blood supply , Brain Neoplasms/drug therapy , Glioblastoma/blood supply , Glioblastoma/drug therapy , Interferon-beta/administration & dosage , Neovascularization, Pathologic/prevention & control , Animals , Biopsy, Needle , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cerebrovascular Circulation , Disease Models, Animal , Disease Progression , Gene Transfer Techniques , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Immunohistochemistry , Infusions, Intralesional , Random Allocation , Rats , Reference Values , Survival Analysis , Transplantation, Heterologous , Xenograft Model Antitumor Assays
4.
Int J Radiat Oncol Biol Phys ; 76(5): 1537-45, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20338480

ABSTRACT

PURPOSE: Ionizing radiation, an important component of glioma therapy, is critically dependent on tumor oxygenation. However, gliomas are notable for areas of necrosis and hypoxia, which foster radioresistance. We hypothesized that pharmacologic manipulation of the typically dysfunctional tumor vasculature would improve intratumoral oxygenation and, thus, the antiglioma efficacy of ionizing radiation. METHODS AND MATERIALS: Orthotopic U87 xenografts were treated with either continuous interferon-beta (IFN-beta) or bevacizumab, alone, or combined with cranial irradiation (RT). Tumor growth was assessed by quantitative bioluminescence imaging; the tumor vasculature using immunohistochemical staining, and tumor oxygenation using hypoxyprobe staining. RESULTS: Both IFN-beta and bevaziumab profoundly affected the tumor vasculature, albeit with different cellular phenotypes. IFN-beta caused a doubling in the percentage of area of perivascular cell staining, and bevacizumab caused a rapid decrease in the percentage of area of endothelial cell staining. However, both agents increased intratumoral oxygenation, although with bevacizumab, the effect was transient, being lost by 5 days. Administration of IFN-beta or bevacizumab before RT was significantly more effective than any of the three modalities as monotherapy or when RT was administered concomitantly with IFN-beta or bevacizumab or 5 days after bevacizumab. CONCLUSION: Bevacizumab and continuous delivery of IFN-beta each induced significant changes in glioma vascular physiology, improving intratumoral oxygenation and enhancing the antitumor activity of ionizing radiation. Additional investigation into the use and timing of these and other agents that modify the vascular phenotype, combined with RT, is warranted to optimize cytotoxic activity.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Glioma/blood supply , Neovascularization, Pathologic/drug therapy , Oxygen Consumption/drug effects , Radiation Tolerance/drug effects , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Bevacizumab , Cell Hypoxia/drug effects , Glioma/metabolism , Glioma/radiotherapy , Interferon-beta/pharmacology , Male , Mice , Mice, SCID , Transplantation, Heterologous
5.
J Neurooncol ; 94(2): 203-12, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19337695

ABSTRACT

INTRODUCTION: We have completed in vivo safety and efficacy studies of the use of a novel drug delivery system, a gel matrix-temozolomide formulation that is injected intracranially into the post-resection cavity, as a candidate for glioma therapy. METHODS: A rat intracranial resection model of C6-GFP intracranial glioma was used for safety and toxicity studies. Biodistribution studies were performed using gel matrix-gallocyanine formulations and were evaluated at various time intervals using real-time analysis of dye distribution. Additionally, the resection model was used to determine the efficacy of gel matrix-temozolomide as compared to blank gel matrix. A subcutaneous human xenograft glioma model was used to further assess the efficacy of gel matrix-temozolomide in reducing the overall tumor load. RESULTS: Gel matrix-temozolomide exhibited minimal cytotoxicity toward normal brain tissue while displaying high levels of oncolytic activity toward glioma cells. In the intracranial glioma resection and subcutaneous glioma model, administration of gel matrix-temozolomide directly to the tumor bed was well tolerated and effective at reducing the tumor load. A significant reduction of tumor load was observed (P < 0.0001) in the 30% temozolomide group (approximately 95%) as compared to blank control. There was little morbidity and no mortality associated with gel matrix treatment. CONCLUSIONS: Gel matrix-temozolomide appears to be safe and effective when used in vivo to treat intracranial glioma and warrants further development as a potential adjuvant therapy.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Biocompatible Materials/administration & dosage , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Disease Models, Animal , Drug Delivery Systems , Glioma/drug therapy , Animals , Antineoplastic Agents, Alkylating/pharmacokinetics , Brain/drug effects , Brain/pathology , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Combined Modality Therapy , Dacarbazine/administration & dosage , Dacarbazine/pharmacokinetics , Drug Implants/administration & dosage , Glioma/pathology , Glioma/surgery , Humans , Maximum Tolerated Dose , Mice , Polymers , Rats , Rats, Wistar , Temozolomide , Tissue Distribution , Tumor Cells, Cultured
6.
J Neurosurg ; 106(2 Suppl): 165-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17330548

ABSTRACT

The authors report on the development of an anterior cerebral artery pseudoaneurysm that hemorrhaged after monopolar coagulation for a ventricular catheter lodged in the interhemispheric fissure. After observing this complication, the authors developed a simple bench test that can be performed by any neurosurgeon to determine the safest coagulation parameters for any given diathermy unit. A modified grounding pad was placed in a beaker of a protein solution consisting of egg whites. Ventricular catheters were then placed in the solution, and a monopolar diathermy current was applied to a metal stylet at various wattages and for different durations of time. Inducing coagulation at 40 W with a diathermy unit produced flames emanating from around the pores of the catheter tip. Flash flames were also observed at 35 W, forming a coagulum of egg white for a distance of up to 1 cm from the catheter tip. All heat was dissipated through the holes of the first 16 mm of the catheter. At 20 W the flame was minimal and coagulation appeared adequate, whereas at 15 W only bubbles were seen around the tip together with suboptimal coagulum formation. This technique is a simple and effective means of determining the optimal setting for monopolar diathermy and can be used to figure the optimal catheter coagulation wattage for a given diathermy unit. Considering the results of this study, the authors have lowered the current for coagulation in ventricular catheters to 20 W.


Subject(s)
Electrocoagulation/instrumentation , Ventriculoperitoneal Shunt/instrumentation , Aneurysm, False/etiology , Aneurysm, False/surgery , Anterior Cerebral Artery/injuries , Cerebral Hemorrhage/etiology , Cerebral Ventricles/surgery , Child , Choroid Plexus/surgery , Electrocoagulation/standards , Equipment Failure , Humans , Intracranial Aneurysm/etiology , Intracranial Aneurysm/surgery , Male , Materials Testing , Subarachnoid Hemorrhage/etiology , Ventriculoperitoneal Shunt/adverse effects
7.
Childs Nerv Syst ; 22(10): 1296-300, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16761160

ABSTRACT

OBJECTIVE: After tumor resection involving the posterior fossa, postoperative swallowing dysfunction may be anticipated. This retrospective study was designed to document swallowing abnormalities in children after posterior fossa brain tumor surgery and to recommend management approaches for children at risk for aspiration. METHODS: Twenty-four children referred postoperatively for a video fluoroscopic swallow study (VFSS) out of 127 children undergoing posterior fossa surgery for brain tumor resection from 1998 to 2001 were evaluated for oral, pharyngeal, and cervical esophageal transits. CONCLUSION: Less than half of the 24 children swallowed functionally within the first week after surgery. VFSS was an appropriate tool for diagnosing swallowing dysfunction, which is impossible to determine at bedside. Children with brain stem involvement and more difficult recovery had significantly worse swallowing function and greater aspiration. We recommend that children with compromised swallowing function should not feed orally until a radiographic swallowing assessment demonstrates functional and safe swallowing with or without therapeutic intervention.


Subject(s)
Deglutition/physiology , Infratentorial Neoplasms/physiopathology , Infratentorial Neoplasms/surgery , Postoperative Period , Adolescent , Child , Child, Preschool , Cohort Studies , Female , Humans , Male , Retrospective Studies
8.
J Neurooncol ; 77(2): 143-52, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16314952

ABSTRACT

OBJECTIVE: To test in vitro and in vivo the safety and efficacy of a novel chemotherapeutic agent, KM-233, for the treatment of glioma. METHODS: In vitro cell cytotoxicity assays were used to measure and compare the cytotoxic effects of KM-233, Delta(8)-tetrahydrocannabinol (THC), and bis-chloroethyl-nitrosurea (BCNU) against human U87 glioma cells. An organotypic brain slice culture model was used for safety and toxicity studies. A human glioma-SCID mouse side-pocket tumor model was used to test in vivo the safety and efficacy of KM-233 with intratumoral and intra-peritoneal administration. RESULTS: KM-233 is a classical cannabinoid with good blood brain barrier penetration that possesses a selective affinity for the CB2 receptors relative to THC. KM-233 was as efficacious in its cytotoxicity against human U87 glioma as Delta(8)-tetrahydrocannabinol, and superior to the commonly used anti-glioma chemotherapeutic agent, BCNU. The cytotoxic effects of KM-233 against human glioma cells in vitro occur as early as two hours after administration, and dosing of KM-233 can be cycled without compromising cytotoxic efficacy and while improving safety. Cyclical dosing of KM-233 to treat U87 glioma in a SCID mouse xenograft side pocket model was effective at reducing the tumor burden with both systemic and intratumoral administration. CONCLUSION: These studies provide both in vitro and in vivo evidence that KM-233 shows promising efficacy against human glioma cell lines in both in vitro and in vivo studies, minimal toxicity to healthy cultured brain tissue, and should be considered for definitive preclinical development in animal models of glioma.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Cannabinoids/pharmacology , Glioma/drug therapy , Animals , Carmustine/pharmacology , Cell Line, Tumor , Dronabinol/pharmacology , Drug Administration Schedule , Humans , Inhibitory Concentration 50 , Mice , Mice, SCID , Organ Culture Techniques
9.
Brain Res ; 1047(1): 56-64, 2005 Jun 14.
Article in English | MEDLINE | ID: mdl-15893739

ABSTRACT

The myelodysplasia/myeloid leukemia factor 1-interacting protein MLF1IP is a novel gene which encodes for a putative transcriptional repressor. It is localized to human chromosome 4q35.1 and is expressed in both the nuclei and cytoplasm of cells. Northern and Western blot analyses have revealed MLF1IP to be present at very low amounts in normal brain tissues, whereas a number of human and rat glioblastoma (GBM) cell lines demonstrated a high level expression of the MLF1IP protein. Immunohistochemical analysis of rat F98 and C6 GBM tumor models showed that MLF1IP was highly expressed in the tumor core where it was co-localized with MLF1 and nestin. Moreover, MLF1IP expression was elevated in the contralateral brain where no tumor cells were detected. These observations, together with previous data demonstrating a role for MLF1IP in erythroleukemias, suggest a possible function for this protein in glioma pathogenesis and potentially in other types of malignancies.


Subject(s)
Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Nuclear Proteins/metabolism , Animals , Brain Neoplasms/physiopathology , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins , Disease Models, Animal , Glioblastoma/physiopathology , Histones , Humans , Immunohistochemistry , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Proteins/metabolism , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Up-Regulation/physiology
10.
J Neurooncol ; 71(3): 245-55, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15735912

ABSTRACT

To investigate adult neural stem cell (NSC) biology in relation to glioma, the C6 glioma cell line was tagged with green fluorescent protein (GFP) and inoculated into the brain of adult rats. The in vivo biological response of the brain to glioma was studied using immunohistochemical analysis of the subventricular zone (SVZ), peritumoral areas, and glioma. Nestin immunoreactive cells were found infiltrating glioma, but the distribution of abnormal immunoreactivity was restricted to the dorsal and medial border of the tumor relative to the ipsilateral ventricle. The SVZ was found to be hypertrophic, hypercellular, and up-regulated nestin expression. Furthermore, a dense contiguous population of nestin immunoreactive cells could be found streaming from ipsilateral dorsal tip of the SVZ, tracking along the ventral margin of the corpus callosum, and fanning out to encompass and infiltrate the proximal tumor border. Although most cells were either nestin or glial fibrillary acidic protein (GFAP) immunoreactive in the SVZ and along the ventral margin of the corpus callosum, the number of cells co-expressing both markers increased proportionally as the tumor was approached so that the predominant cell population along the proximal tumor border was GFAP immunoreactive. Finally, we demonstrated that a significant proportion of cells found in areas of abnormal immunoreactivity were proliferating, especially in peritumoral areas. In summary, there is an induction of neuropoietic activity in a rat intracranial glioma model that results in an infiltration and accumulation of abnormal nestin and GFAP expressing cells with proliferative potential along the dorsal and medial border of intracranial C6 glioma.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioma/metabolism , Glioma/pathology , Neuroglia/metabolism , Neuroglia/pathology , Stem Cells/metabolism , Animals , Cell Differentiation , Cell Lineage , Cell Movement , Cerebral Ventricles/pathology , Disease Models, Animal , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Intermediate Filament Proteins/metabolism , Neoplasm Transplantation , Nerve Tissue Proteins/metabolism , Nestin , Rats , Rats, Inbred WF , Rats, Sprague-Dawley , Stem Cells/cytology , Up-Regulation
11.
Cancer Cell ; 7(1): 87-99, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15652752

ABSTRACT

Activation of Stat5 is frequently found in leukemias. To study the mechanism and role of Stat5 activation, we introduced a constitutively activated Stat5a mutant, cS5F, into murine bone marrow (BM) cells. BM transplantation with cS5F-transfected cells caused development of multilineage leukemias in lethally irradiated wild-type or nonirradiated Rag2(-/-) mice. The leukemic cells showed strongly enhanced levels of cS5F tetramers but unchanged cS5F dimer levels in a DNA binding assay. Moreover, Stat5a mutants engineered to form only dimers, but not tetramers, failed to induce leukemias. In addition, Stat5 tetramers were found to accumulate in excess compared to dimers in various human leukemias. These data suggest that Stat5 tetramers are associated with leukemogenesis.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Leukemia/metabolism , Milk Proteins/chemistry , Milk Proteins/metabolism , Protein Structure, Quaternary , Trans-Activators/chemistry , Trans-Activators/metabolism , Animals , Biomarkers , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Bone Marrow Transplantation , Cell Lineage , Cell Transformation, Neoplastic , Cells, Cultured , DNA-Binding Proteins/genetics , Female , Genetic Complementation Test , Growth Substances/metabolism , Humans , Leukemia/genetics , Leukemia/pathology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Milk Proteins/genetics , Mutation , Nuclear Proteins , Oncogenes , STAT5 Transcription Factor , Spleen/metabolism , Spleen/pathology , Trans-Activators/genetics , Transfection , Tumor Suppressor Proteins
12.
J Neurooncol ; 70(1): 3-15, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15527101

ABSTRACT

Cadherins are Ca2+-dependent cell adhesion molecules that play an important role in tissue construction and morphogenesis in multicellular organisms. Over the last few years, reports have emerged in the literature describing the involvement of cadherins in tumor invasion and metastasis. Cadherins typically demonstrate up and down-regulation according to the biological needs of the tissue. Additionally, up-regulation of N-cadherin is thought to be important for tumor formation in early stages of tumor development. We studied N-cadherin in surgical specimens of patients with primary glioblastoma by microarray analysis and found that N-cadherin mRNA expression is up-regulated compared to normal brain. To study the effects of N-cadherin expression on invasion and metastasis in vitro and in vivo, we overexpressed N-cadherin in the rat C6 glioma cell line which normally has low levels of N-cadherin. We found that up-regulation of N-cadherin resulted in a slight decreased adhesion to type IV collagen, fibronectin, and laminin, but statistically significant decreased adhesion to type I collagen. Furthermore, increased expression of N-cadherin correlated with a dramatic decrease in invasive behavior in extracellular matrix invasion assays. We then proceeded to study these cell lines in vivo in a rat intracranial glioma model, and found that N-cadherin expression inversely correlated with invasion into surrounding tissues, irregular margins, and extracranial invasion. In summary, these data collectively demonstrate that N-cadherin levels are important in the malignant behavior of gliomas, and may serve as a prognostic indicator for patients with high-grade gliomas.


Subject(s)
Brain Neoplasms/metabolism , Cadherins/metabolism , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Adult , Aged , Aged, 80 and over , Animals , Brain Neoplasms/pathology , Cadherins/genetics , Cell Adhesion , Cell Aggregation/drug effects , Collagen Type I/metabolism , Collagen Type IV/metabolism , Female , Fibronectins/metabolism , Gene Expression Profiling , Glioma/pathology , Humans , Laminin/metabolism , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured
13.
J Neurosurg ; 100(6): 1049-59, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15200120

ABSTRACT

OBJECT: The purpose of this study was to evaluate both replication-competent and replication-restricted recombinant vesicular stomatitis virus (VSV) vectors as therapeutic agents for high-grade gliomas by using an organotypic brain tissue slice-glioma coculture system. METHODS: The coculture system involved growing different brain structures together to allow neurons from these tissues to develop synaptic connections similar to those found in vivo. Rat C6 or human U87 glioma cells were then introduced into the culture to evaluate VSV as an oncolytic therapy. The authors found that recombinant wild-type VSV (rVSV-wt) rapidly eliminated C6 glioma cells from the coculture, but also caused significant damage to neurons, as measured by a loss of microtubule-associated protein 2 immunoreactivity and a failure in electrophysiological responses from neurons in the tissue slice. Nonetheless, pretreatment with interferon beta (IFNbeta) virtually eliminated VSV infection in healthy tissues without impeding any oncolytic effects on tumor cells. Despite the protective effects of the IFNbeta pretreatment, the tissue slices still showed signs of cytopathology when exposed to rVSV-wt. In contrast, pretreatment with IFNbeta and inoculation with a replication-restricted vector with its glycoprotein gene deleted (rVSV-deltaG) effectively destroyed rat C6 and human U87 glioma cells in the coculture, without causing detectable damage to the neuronal integrity and electrophysiological properties of the healthy tissue in the culture. CONCLUSIONS: Data in this study provide in vitro proof-of-principle that rVSV-deltaG is an effective oncolytic agent that has minimal toxic side effects to neurons compared with rVSV-wt and therefore should be considered for development as an adjuvant to surgery in the treatment of glioma.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/pathogenicity , Animals , Brain Neoplasms/pathology , Culture Techniques , Electrophysiology , Glioma/pathology , Humans , Interferon-beta/pharmacology , Neurons , Rats , Synapses , Tumor Cells, Cultured
14.
EMBO J ; 21(4): 653-64, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11847113

ABSTRACT

TACC3 is a centrosomal/mitotic spindle-associated protein that is highly expressed in a cell cycle-dependent manner in hematopoietic lineage cells. During embryonic development, TACC3 is expressed in a variety of tissues in addition to the hematopoietic lineages. TACC3 deficiency causes an embryonic lethality at mid- to late gestation involving several lineages of cells. Hematopoietic stem cells, while capable of terminal differentiation, are unable to be expanded in vitro or in vivo in reconstitution approaches. Although gross alterations in centrosome numbers and chromosomal segregation are not observed, TACC3 deficiency is associated with a high rate of apoptosis and expression of the p53 target gene, p21(Waf1/Cip1). Hematopoietic stem cell functions, as well as deficiencies in other cell lineages, can be rescued by combining the TACC3 deficiency with p53 deficiency. The results support the concept that TACC3 is a critical component of the centrosome/mitotic spindle apparatus and its absence triggers p53-mediated apoptosis.


Subject(s)
Apoptosis/physiology , Centrosome/metabolism , Hematopoietic Stem Cells/physiology , Microtubule-Associated Proteins/physiology , Tumor Suppressor Protein p53/physiology , Animals , Base Sequence , Blotting, Northern , DNA Primers , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Genes, Lethal , In Situ Nick-End Labeling , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...