Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Oncogene ; 31(10): 1264-74, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-21804599

ABSTRACT

The phosphatidylinositol-3-OH kinase (PI3K)-Akt pathway is activated in cancer by genetic or epigenetic events and efforts are under way to develop targeted therapies. phosphatase and tensin homolog deleted on chromosome 10 (PTEN) tumor suppressor is the major brake of the pathway and a common target for inactivation in glioblastoma, one of the most aggressive and therapy-resistant cancers. To achieve potent inhibition of the PI3K-Akt pathway in glioblastoma, we need to understand its mechanism of activation by investigating the interplay between its regulators. We show here that PTEN modulates the PI3K-Akt pathway in glioblastoma within a tumor suppressor network that includes Na(+)/H(+) exchanger regulatory factor 1 (NHERF1) and pleckstrin-homology domain leucine-rich repeat protein phosphatases 1 (PHLPP1). The NHERF1 adaptor, previously characterized by our group as a PTEN ligand and regulator, shows also PTEN-independent Akt-modulating effects that led us to identify the PHLPP1/PHLPP2 Akt phosphatases as NHERF1 ligands. NHERF1 interacts via its PDZ domains with PHLPP1/PHLPP2 and scaffolds heterotrimeric complexes with PTEN. Functionally, PHLPP1 requires NHERF1 for membrane localization and growth-suppressive effects. PHLPP1 loss boosts Akt phosphorylation only in PTEN-negative cells and cooperates with PTEN loss for tumor growth. In a panel of low-grade and high-grade glioma patient samples, we show for the first time a significant disruption of all three members of the PTEN-NHERF1-PHLPP1 tumor suppressor network in high-grade tumors, correlating with Akt activation and patient's abysmal survival. We thus propose a PTEN-NHERF1-PHLPP PI3K-Akt pathway inhibitory network that relies on molecular interactions and can undergo parallel synergistic hits in glioblastoma.


Subject(s)
Brain Neoplasms/etiology , Glioblastoma/etiology , Nuclear Proteins/physiology , PTEN Phosphohydrolase/physiology , Phosphoprotein Phosphatases/physiology , Phosphoproteins/physiology , Sodium-Hydrogen Exchangers/physiology , Tumor Suppressor Proteins/physiology , Aged , Humans , Middle Aged , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction
2.
Oncogene ; 29(31): 4460-72, 2010 Aug 05.
Article in English | MEDLINE | ID: mdl-20531304

ABSTRACT

Angiogenesis involves a complex set of cell-cell and cell-extracellular matrix (ECM) interactions that coordinately promote and inhibit blood vessel growth and sprouting. Although many factors that promote angiogenesis have been characterized, the identities and mechanisms of action of endogenous inhibitors of angiogenesis remain unclear. Furthermore, little is known about how cancer cells selectively circumvent the actions of these inhibitors to promote pathological angiogenesis, a requisite event for tumor progression. Using mosaic mouse models of the malignant brain cancer, astrocytoma, we report that tumor cells induce pathological angiogenesis by suppressing expression of the ECM protein receptor alphavbeta8 integrin. Diminished integrin expression in astrocytoma cells leads to reduced activation of latent TGFbetas, resulting in impaired TGFbeta receptor signaling in tumor-associated endothelial cells. These data reveal that astrocytoma cells manipulate their angiogenic balance by selectively suppressing alphavbeta8 integrin expression and function. Finally, these results show that an adhesion and signaling axis normally involved in developmental brain angiogenesis is pathologically exploited in adult brain tumors.


Subject(s)
Astrocytoma/blood supply , Astrocytoma/genetics , Brain Neoplasms/blood supply , Brain Neoplasms/genetics , Integrins/physiology , Neovascularization, Pathologic/genetics , Animals , Animals, Newborn , Astrocytoma/pathology , Brain Neoplasms/pathology , Disease Models, Animal , Down-Regulation , Humans , Integrins/genetics , Male , Mice , Mice, Nude , Mice, Transgenic , Mosaicism , Neoplasm Transplantation , Neovascularization, Pathologic/prevention & control , Transplantation, Heterologous , Tumor Cells, Cultured
3.
Cancer Gene Ther ; 16(3): 256-65, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18927600

ABSTRACT

The fact that glioblastomas, which are one of the most devastating cancers, frequently express the Delta-EGFR (epithelial growth factor receptor) also called mutant variant III of EGFR (EGFRvIII) suggests that this cancer cell-specific receptor might serve as an ideal target for cancer therapy. To assess its potential as such a target, we constructed an oncolytic adenovirus with Retargeted Infectivity Via EGFR (Delta-24-RIVER) on the backbone of Delta-24. This new oncolytic adenovirus targets, as Delta-24 does, the disrupted Rb pathway in cancer cells; in addition, this adenovirus has also been retargeted through the abrogation of CAR binding (Y477A mutation in adenoviral fiber protein) and insertion of an EGFRvIII-specific binding peptide in the HI loop of the fiber protein. As compared with Delta-24, Delta-24-RIVER induced EGFRvIII-selective cytotoxicity in U-87 MG isogenic cell lines and in tetracycline-inducible EGFRVIII expressing U-251 MG cells. Accordingly, by tittering the viral progeny and examining fiber protein expression in the above cells, we showed that the replication of this new construct also correlated with EGFRvIII expression. Consistently, immunohistochemistry staining of the adenoviral capsid protein hexon in the virus-treated tumors revealed that the virus replicated more efficiently in EGFRvIII-expressing U-87 MG.DeltaEGFR xenografts than in the tumors grown from U-87 MG cells. Importantly, treatment with Delta-24-RIVER prolonged the survival of animals with intracranial xenografts derived from U-87 MG.DeltaEGFR cells. Therefore, our results constitute the first proof of the direct targeting of a cancer-specific receptor using an oncolytic adenovirus.


Subject(s)
Adenoviruses, Human/physiology , Brain Neoplasms/therapy , ErbB Receptors/antagonists & inhibitors , Genetic Vectors/therapeutic use , Glioblastoma/therapy , Neoplasm Proteins/antagonists & inhibitors , Oncolytic Virotherapy , Adenoviruses, Human/genetics , Amino Acid Sequence , Animals , Brain Neoplasms/pathology , Cell Line, Tumor/transplantation , Exons/genetics , Genes, Retinoblastoma , Genes, erbB-1 , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Proteins/genetics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Sequence Deletion , Virus Replication
4.
Ann Oncol ; 19(7): 1242-1248, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18334512

ABSTRACT

BACKGROUND: The purpose of this retrospective study was to determine, in a cohort of patients with breast cancer and central nervous system (CNS) metastases, the effect of trastuzumab in patients with human epidermal growth factor receptor 2 (HER2)-positive disease and to compare this with that of patients with HER2-negative disease. METHODS: Five hundred and ninety-eight patients with invasive breast cancer, CNS metastases and known HER2 status were identified. Time to CNS metastases and survival after CNS metastases were estimated by the Kaplan-Meier method, and Cox models were fitted to determine the association between HER2 status, trastuzumab treatment and outcomes after adjustment for other patient characteristics. RESULTS: In the multivariable model, patients with HER2-negative disease [Hazard ratio (HR) 1.50, 95% confidence interval (CI) 1.15-1.95, P = 0.003] and patients with HER2-positive disease who did not receive trastuzumab (HR 2.13, 95% CI 1.51-3.00, P < 0.0001) had shorter times to CNS metastases compared with patients with HER2-positive disease who had received trastuzumab as first-line therapy for metastases. Furthermore, patients with HER2-negative disease (HR 1.66, 95% CI 1.31-2.12, P < 0.0001) and patients with HER2-positive disease who had never received trastuzumab (HR 1.34, 95% CI 0.78-2.30, P = 0.28) had an increased hazard of death compared with patients with HER2-positive disease who had received trastuzumab before or at the time of CNS metastases diagnosis. CONCLUSION: In our cohort of patients with breast cancer and CNS metastases, patients with HER2-positive disease treated with trastuzumab had longer times to development of and better survival from CNS metastases compared with patients with HER2-positive disease who had never received trastuzumab and patients with HER2-negative breast cancer.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Central Nervous System Neoplasms/secondary , Receptor, ErbB-2/metabolism , Adult , Aged , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/therapy , Cohort Studies , Female , Follow-Up Studies , Humans , Middle Aged , Multivariate Analysis , Neoplasm Metastasis/diagnosis , Neoplasm Metastasis/pathology , Neoplasm Metastasis/therapy , Prognosis , Receptor, ErbB-2/genetics , Retrospective Studies , Survival Analysis , Time Factors , Trastuzumab , Treatment Outcome
5.
Oncogene ; 26(42): 6212-9, 2007 Sep 13.
Article in English | MEDLINE | ID: mdl-17404569

ABSTRACT

We recently showed that FoxM1 is overexpressed in human glioblastomas and that forced FoxM1B expression in anaplastic astrocytoma cells leads to the formation of highly invasive glioblastoma multiforme (GBM) in nude mice. However, the molecular mechanisms by which FoxM1 enhances glioma invasion are unknown. In this study, we found that FoxM1 overexpression increased matrix metalloproteinase (MMP)-2 expression in glioma cells, whereas blockade of FoxM1 expression suppressed MMP-2 expression. Transfection of FoxM1 into glioma cells directly activated the MMP-2 promoter, whereas inhibition of FoxM1 expression by FoxM1-siRNA suppressed its activation. We identified a FoxM1-binding site in the MMP-2 promoter and demonstrated that FoxM1 protein bound directly to it. Mutation of this FoxM1-binding site significantly attenuated MMP-2 promoter activity. Furthermore, FoxM1 overexpression increased the invasiveness of glioma cells, whereas inhibition of FoxM1 expression suppressed the invasiveness of GBM cells. Inhibition of MMP-2 by a specific MMP-2 inhibitor reversed the invasive phenotype of glioma cells overexpressing FoxM1. Finally, immunohistochemical analysis of 45 human GBM specimens showed a significant correlation between FoxM1 overexpression and elevated MMP-2 expression. Collectively, these findings provide evidence that FoxM1 contributes to glioma progression by enhancing MMP-2 gene transcription and thus tumor-cell invasion.


Subject(s)
Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Glioma/pathology , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Up-Regulation/physiology , Cell Line, Tumor , Forkhead Box Protein M1 , Forkhead Transcription Factors/physiology , Glioma/enzymology , Glioma/genetics , Humans , Neoplasm Invasiveness , Transcription, Genetic/physiology
6.
Neurology ; 66(1): 127-30, 2006 Jan 10.
Article in English | MEDLINE | ID: mdl-16401863

ABSTRACT

Using whole genome expression microarray technology to discover clinically relevant biomarkers for pilocytic astrocytoma (PA), the authors identified matrilin-2 as a unique mRNA overexpressed in PA. Matrilin-2 protein expression was similarly elevated in the majority of sporadic PA, but in only one neurofibromatosis 1-associated PA with an unusually aggressive clinical phenotype. These results suggest that matrilin-2 may be a specific and clinically useful biomarker for discriminating between indolent and clinically aggressive PA.


Subject(s)
Astrocytoma/diagnosis , Astrocytoma/genetics , Biomarkers, Tumor/genetics , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Extracellular Matrix Proteins/genetics , Glycoproteins/genetics , Astrocytoma/classification , Cell Transformation, Neoplastic/genetics , Child , Chromosomes, Human, Pair 8/genetics , DNA Mutational Analysis , Extracellular Matrix Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Genetic Testing , Glycoproteins/metabolism , Humans , Immunohistochemistry , Matrilin Proteins , Neurofibromatosis 1/complications , Neurofibromatosis 1/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , RNA, Messenger/genetics , Up-Regulation/genetics
7.
Neuropathol Appl Neurobiol ; 29(6): 584-95, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14636165

ABSTRACT

Haematogenous leucocytes enter the central nervous system (CNS) during diverse disorders of varied aetiologies. Understanding the trafficking cues that mediate CNS leucocyte infiltration might promote the development of flexible and selective means to modulate inflammation to achieve clinical benefit. The trafficking machinery of leucocytes has been elucidated during the past decade and consists of cell-surface adhesion molecules, chemoattractant cytokines (chemokines) and their receptors. Recent work in our laboratory characterized chemokine receptors found on T lymphocytes and monocytes in brain sections from subjects with one pathological subtype of multiple sclerosis (MS), an immune-mediated inflammatory demyelinating disease. In these tissues, the types 1 and 5 CC chemokine receptors (CCR1 and CCR5) were detected on perivascular monocytic cells whereas only CCR5 was present on parenchymal macrophages. The type 3 CXC chemokine receptor (CXCR3) was present on virtually all CD3-positive T cells. In the current study, we evaluated the expression of these receptors on the infiltrating cells present in cases of other inflammatory CNS disorders including those of dysimmune, infectious, neoplastic, and vascular aetiology. Perivascular and parenchymal monocytic cells expressed CCR1 in all cases and CXCR3 was consistently present on a substantial proportion of CD3+ T cells. The occurrence of CCR5 on parenchymal macrophages was much less uniform across the varied disorders. These data implicate CCR1 in monocyte infiltration of the CNS and are consistent with reports of studies in CCR1-deficient mice. CXCR3 is also likely to play a role in accumulation of T cells in the inflamed CNS. By contrast, our findings suggest that regulation of CCR5 on phagocytic macrophages may be contingent on the lesion environment.


Subject(s)
Brain Diseases/pathology , Inflammation/pathology , Leukocytes, Mononuclear/metabolism , Receptors, Chemokine/metabolism , Adolescent , Adult , Aged , Brain Diseases/immunology , CD3 Complex/metabolism , Calgranulin B/metabolism , Child , Humans , Immunohistochemistry , Inflammation/immunology , Male , Middle Aged , Retrospective Studies
8.
Int J Radiat Oncol Biol Phys ; 51(2): 410-8, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11567815

ABSTRACT

PURPOSE: Recent studies have suggested relative radioresistance in glioblastoma multiforme (GM) tumors in older patients, consistent with their shorter survival. Two common molecular genetic abnormalities in GM are age related: epidermal growth factor receptor (EGFR) overexpression in older patients and p53 mutations in younger patients. We tested whether these abnormalities correlated with clinical heterogeneity in GM response to radiation treatment. METHODS AND MATERIALS: Radiographically assessed radiation response (5-level scale) was correlated with EGFR immunoreactivity, p53 immunoreactivity, and p53 exon 5-8 mutation status in 170 GM patients treated using 2 prospective clinical protocols. Spearman rank correlation and proportional-odds ordinal regression were used for univariate and multivariate analysis. RESULTS: Positive EGFR immunoreactivity predicted poor radiographically assessed radiation response (p = 0.046). Thirty-three percent of tumors with no EGFR immunoreactivity had good radiation responses (>50% reduction in tumor size by CT or MRI), compared to 18% of tumors with intermediate EGFR staining and 9% of tumors with strong staining. There was no significant relationship between p53 immunoreactivity or mutation status and radiation response. Significant relationships were noted between EGFR score and older age and between p53 score or mutation status and younger age. CONCLUSION: The observed relative radioresistance of some GMs is associated with overexpression of EGFR.


Subject(s)
Brain Neoplasms/radiotherapy , ErbB Receptors/metabolism , Genes, p53/genetics , Glioblastoma/radiotherapy , Neoplasm Proteins/metabolism , Supratentorial Neoplasms/radiotherapy , Adolescent , Adult , Age Factors , Aged , Analysis of Variance , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Male , Middle Aged , Odds Ratio , Prospective Studies , Radiotherapy Dosage , Regression Analysis , Supratentorial Neoplasms/genetics , Supratentorial Neoplasms/metabolism
9.
Cancer Res ; 61(18): 6674-8, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11559533

ABSTRACT

Human malignant gliomas are thought to develop as the result of stepwise accumulations of multiple genetic alterations. Recently, we showed that E6/E7-mediated inactivation of p53/pRb, ras pathway activation (initiated by expression of mutant H-Ras), and expression of human telomerase reverse transcriptase (hTERT) in combination converted normal human astrocytes into cells that formed intracranial tumors resembling human anaplastic astrocytoma (AA). In this study, we created human astrocytes that, in addition to expressing E6/E7, hTERT, and Ras, also expressed a constitutive activated form of Akt intended to mimic the Akt activation noted in grade IV glioblastoma multiforme (GBM). Although these cells grew no differently than astrocytes expressing E6, E7, and H-Ras in vitro or in the first 28 days following s.c. implantation, they ultimately formed tumors four to six times larger than those formed by the E6/E7/hTERT/Ras cells. Unlike the poorly vascularized, necrosis-free AA formed by E6/E7/hTERT/Ras cells, the tumors formed by s.c. or intracranial injection of Akt-expressing cells had large areas of necrosis surrounded by neovascularization and were consistent in appearance with grade IV human GBM. These results show that activation of the Akt pathway is sufficient to allow conversion of human AA to human GBM.


Subject(s)
Astrocytoma/enzymology , Astrocytoma/pathology , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Glioblastoma/enzymology , Glioblastoma/pathology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , Animals , Astrocytes/enzymology , Astrocytes/pathology , Astrocytes/physiology , Astrocytoma/genetics , Brain Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Progression , Enzyme Activation , Glioblastoma/genetics , Humans , Mice , Mice, Nude , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Rats , Signal Transduction/physiology , Transfection
10.
Cancer Res ; 61(13): 4956-60, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11431323

ABSTRACT

The formation of human malignant gliomas is thought to involve the accumulation of multiple genetic alterations. To define the function of specific alterations in glioma formation, we serially introduced genetic alterations functionally equivalent to those noted in human malignant gliomas into normal human astrocytes (NHAs). We then monitored the ability of each of these alterations to contribute to the growth of otherwise genetically stable NHAs into intracranial malignant gliomas. Using this model, we show that expression of human telomerase catalytic component (hTERT), but not E7-mediated inactivation of pRb or E6/E7-mediated inactivation of p53/pRb, was sufficient to initiate the tumorigenic process by circumventing cellular senescence in astrocytes. hTERT expression, even in combination with inactivation of p53/pRb, did not transform astrocytes. These alterations together, however, cooperated with ras pathway activation (initiated by expression of mutant H-Ras), but not with phosphatidylinositol 3-kinase pathway activation (initiated by expression of myristoylated Akt) or epidermal growth factor receptor activation, to allow for the formation of intracranial tumors strongly resembling p53/pRb pathway-deficient, telomerase-positive, ras-activated human grade III anaplastic astrocytomas. These results identify four pathways as key in the development of human anaplastic astrocytomas.


Subject(s)
Astrocytes/physiology , Astrocytoma/genetics , Brain Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , RNA , Repressor Proteins , Animals , Astrocytes/pathology , DNA-Binding Proteins , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Oncogene Proteins, Viral/biosynthesis , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/physiology , Papillomavirus E7 Proteins , Retinoblastoma Protein/antagonists & inhibitors , Signal Transduction/genetics , Telomerase/biosynthesis , Telomerase/genetics , Telomerase/physiology , Transfection , Tumor Suppressor Protein p53/antagonists & inhibitors , ras Proteins/physiology
12.
Proc Natl Acad Sci U S A ; 97(9): 4914-9, 2000 Apr 25.
Article in English | MEDLINE | ID: mdl-10781098

ABSTRACT

Acetylcholine excites many central and autonomic neurons through inhibition of M-channels, slowly activating, noninactivating voltage-gated potassium channels. We here provide information regarding the in vivo distribution and biochemical characteristics of human brain KCNQ2 and KCNQ3, two channel subunits that form M-channels when expressed in vitro, and, when mutated, cause the dominantly inherited epileptic syndrome, benign neonatal familial convulsions. KCNQ2 and KCNQ3 proteins are colocalized in a somatodendritic pattern on pyramidal and polymorphic neurons in the human cortex and hippocampus. Immunoreactivity for KCNQ2, but not KCNQ3, is also prominent in some terminal fields, suggesting a presynaptic role for a distinct subgroup of M-channels in the regulation of action potential propagation and neurotransmitter release. KCNQ2 and KCNQ3 can be coimmunoprecipitated from brain lysates. Further, KCNQ2 and KCNQ3 are coassociated with tubulin and protein kinase A within a Triton X-100-insoluble protein complex. This complex is not associated with low-density membrane rafts or with N-methyl-d-aspartate receptors, PSD-95 scaffolding proteins, or other potassium channels tested. Our studies thus provide a view of a signaling complex that may be important for cognitive function as well as epilepsy. Analysis of this complex may shed light on the unknown transduction pathway linking muscarinic acetylcholine receptor activation to M-channel inhibition.


Subject(s)
Brain/metabolism , Epilepsy/genetics , Potassium Channels/analysis , Potassium Channels/genetics , Brain/pathology , Cell Line , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Epilepsy/pathology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunohistochemistry , KCNQ2 Potassium Channel , KCNQ3 Potassium Channel , Macromolecular Substances , Potassium Channels, Voltage-Gated , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis , Subcellular Fractions/metabolism , Transfection
13.
Cancer Res ; 58(17): 3819-25, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9731490

ABSTRACT

Surgical resection followed by radiation therapy is the mainstay of treatment for glioblastoma multiforme (GBM), the most aggressive of the malignant gliomas. The poor clinical response of GBM and the intrinsic radiation resistance of this tumor type have prompted clinical investigations seeking to define the role of chemotherapy in the treatment of GBM. In this study, we examined the cytotoxic response of GBM-derived cell lines to treatment with both radiation and chemotherapy. We observed that the sensitivity of glioma cells to cisplatin- and FAS-induced apoptosis was diminished by prior treatment with ionizing radiation. Radiation conferred resistance to cisplatin and FAS cytotoxicity in a dose- and time-dependent manner. Radiation diminished the cisplatin-induced cytotoxicity of malignant glioma cells but failed to alter the cisplatin susceptibility of normal primary human astrocytes. Given the role of p53 in the response of cells to irradiation, we evaluated whether p53 function affects the observed radiation-induced resistance to cisplatin. By examining isogenic cell lines differing only in p53 function, we demonstrated that radiation conferred resistance to cisplatin independently of p53. Current clinical strategies in the treatment of astrocytic tumors, which include combined modality therapy, have been empirically derived from limited clinical experience. Further understanding of the molecular determinants of apoptosis associated with combined modality therapy may guide the design of more efficacious multimodality protocols.


Subject(s)
Apoptosis/radiation effects , Glioblastoma/therapy , Apoptosis/drug effects , Cisplatin/therapeutic use , Combined Modality Therapy , G1 Phase , Glioblastoma/pathology , Humans , Tumor Cells, Cultured , fas Receptor/physiology
14.
J Magn Reson Imaging ; 8(4): 799-806, 1998.
Article in English | MEDLINE | ID: mdl-9702880

ABSTRACT

To compare the performance of macromolecular albumin gadolinium-diethylenetriamine pentaacetic acid (Gd-DTPA)30 and low molecular weight gadopentetate dimeglumine for microvessel characterization, we examined an intracranial 9L glioma model in which increased angiogenesis, hypervascularity, and hyperpermeability mimic characteristics of clinical malignant brain tumors. Dynamic MRI data were analyzed using a bidirectional, two-compartment kinetic model to extract quantitative estimates for fractional blood volume (fBV) and permeability surface area product (PS). Three criteria were used for comparison of contrast agent performance: (a) tumor conspicuity, defined as the contrast-to-noise ratio (CNR); (b) dynamic range of differential permeability estimates between tumor and normal brain; (c) reasonableness of blood volume estimates. Gadopentetate was superior to macromolecular albumin-(Gd-DTPA)30 for detection of 9L brain gliomas and for measurements of hyperpermeability.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Gadolinium DTPA , Glioma/pathology , Magnetic Resonance Imaging , Albumins , Animals , Brain/blood supply , Brain Neoplasms/blood supply , Contrast Media , Glioma/blood supply , Microcirculation/physiology , Rats , Rats, Inbred F344
15.
Cancer Res ; 57(17): 3635-9, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9288761

ABSTRACT

To gain insight into p53 tissue-specific regulatory pathways and biological activities, we investigated mechanisms that may account for the elevated levels of p53 protein in human foreskin keratinocytes, relative to levels in dermal fibroblasts in vitro. Here, we report that the loss of cell anchorage resulted in an approximately 5-fold decrease in p53 levels in keratinocytes, which was reversible upon reattachment of cells to a substratum. In contrast, fibroblasts did not exhibit such adhesion-dependent regulation of p53 protein. Furthermore, p53 function was attenuated in keratinocytes relative to fibroblasts. These results link p53 to cell adhesion pathways and may provide a molecular basis for epigenetic differences in the maintenance of genomic stability among normal cell types.


Subject(s)
Cell Adhesion/physiology , Keratinocytes/metabolism , Nuclear Proteins , Tumor Suppressor Protein p53/metabolism , Cell Cycle/genetics , Cell Nucleus/metabolism , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , DNA Damage , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Keratinocytes/radiation effects , Male , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , RNA, Messenger/metabolism , Transcriptional Activation/radiation effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/radiation effects
16.
J Neurooncol ; 31(1-2): 17-23, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9049826

ABSTRACT

We screened human primary and recurrent malignant glioma, juvenile pilocytic astrocytoma, medulloblastoma, and meningioma tissue specimens for alterations in p16 gene structure. Single strand conformation polymorphism (SSCP) analysis was used to screen for point mutations, and a quantitative polymerase chain reaction-based assay was used to screen for homozygous gene deletions. In malignant glioma specimens, homozygous p16 gene deletions were significantly more common in high-grade tumors than in low-grade gliomas. Point mutations causing alteration in predicted protein structure were not detected. Medulloblastomas showed rare homozygous deletions and no point mutations. No mutations were detected in meningiomas.


Subject(s)
Carrier Proteins/genetics , Cerebellar Neoplasms/genetics , Enzyme Inhibitors/metabolism , Medulloblastoma/genetics , Meningeal Neoplasms/genetics , Meningioma/genetics , Cyclin-Dependent Kinase Inhibitor p16 , DNA Mutational Analysis , Gene Deletion , Genes, Tumor Suppressor/genetics , Humans , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational
17.
Cancer Res ; 55(4): 727-30, 1995 Feb 15.
Article in English | MEDLINE | ID: mdl-7531613

ABSTRACT

The nitric oxide synthases (NOS) are a family of related enzymes which regulate the production of NO, a free radical gas implicated in a wide variety of biological processes. Vasodilation and increased tumor blood flow, increased vascular permeability, modulation of host tumoricidal activity, and free radical injury to tumor cells and adjacent normal tissues are pathophysiological features of malignant tumors that may be mediated by NO. We examined human brain tumors for three NOS isoforms and NADPH diaphrase, a histochemical marker of NOS activity in the brain. We detected increased expression of the brain and endothelial forms of NOS [NOS I and NOS II, respectively (C. Nathan and Q. Xie. Cell, 78: 915-919, 1994)] in astrocytic tumors, and the highest levels of expression was found in higher grade tumors. Each of these two isoforms was found in tumor cells and tumor endothelial cells. The macrophage isoform of NOS (NOS III) was less frequently detected and expressed at a lower level, predominantly in tumor endothelial cells. NADPH diaphorase staining for NOS activity paralleled this pattern of NOS expression. Western blot analysis of tumor tissues for these NOS isoforms confirmed these observations. Our data indicate that malignant central nervous system neoplasms express unexpectedly high levels of NOS and suggest that NO production may be associated with pathophysiological processes important to these tumors.


Subject(s)
Amino Acid Oxidoreductases/analysis , Brain Neoplasms/enzymology , Isoenzymes/analysis , Blotting, Western , Brain/enzymology , Brain Neoplasms/chemistry , Brain Neoplasms/pathology , Cell Division/physiology , Glioblastoma/chemistry , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Immunohistochemistry , NADPH Dehydrogenase/analysis , Nitric Oxide Synthase
18.
Blood ; 79(7): 1768-74, 1992 Apr 01.
Article in English | MEDLINE | ID: mdl-1373087

ABSTRACT

The majority of lymphomas in the setting of acquired, iatrogenic, or congenital immunodeficiencies are B-cell lymphoproliferations. We describe a rare T-cell lymphoma in a fulminantly ill patient infected with human immunodeficiency virus type 1 (HIV-1). The T-cell nature of the process was defined genotypically (monoclonal T-cell receptor beta-chain [CT beta] rearrangement) and phenotypically (CD45RO+, CD4+, CD5+, CD25+, CD8-, CD3- and negative for a variety of B-cell and monocyte markers). The CD4+, CD25+ (interleukin-2 receptor [IL-2R]) phenotype with production of IL-2 and IL-2R RNA is analogous to human T-lymphotropic virus type I (HTLV-I)-associated adult T-cell leukemia/lymphoma (ATLL); however, no HTLV-1 could be detected. Southern blot analysis did demonstrate monoclonally integrated HIV-1 within the tumor genome. Furthermore, the tumor cells were producing HIV p24 antigen as shown by immunohistochemistry. This is the first case of acquired immunodeficiency syndrome (AIDS)-associated non-Hodgkin's lymphoma in which HIV-1 infection may have played a central role in the lymphocyte transformation process.


Subject(s)
Acquired Immunodeficiency Syndrome/complications , Cell Transformation, Viral , HIV-1 , Lymphoma, T-Cell/etiology , T-Lymphocytes , Base Sequence , Blotting, Southern , CD4-Positive T-Lymphocytes/pathology , DNA, Viral/analysis , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Human T-lymphotropic virus 1/genetics , Humans , Immunophenotyping , Interleukin-2/genetics , Lung/pathology , Lymph Nodes/pathology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/microbiology , Male , Middle Aged , Molecular Sequence Data , RNA/metabolism , T-Lymphocytes/immunology
19.
Am J Clin Pathol ; 95(4): 499-502, 1991 Apr.
Article in English | MEDLINE | ID: mdl-2014775

ABSTRACT

Cerebral phaeohyphomycosis (also referred to as cerebral chromomycosis), one of the diseases caused by the dematiaceous (black) fungi, is most commonly caused by Cladosporium trichoides (referred to by some as Xylohypha bantiana) and is a rare disease, with 31 culture-proven cases reported to date. Although most cases have occurred in immunocompetent hosts, recent experimental evidence suggests that host immunosuppression may predispose patients to the disease. The authors report a case of fatal cerebral phaeohyphomycosis in a liver transplant patient, the first to occur in a transplant patient of any type, to the best of the authors' knowledge. This case provides support for the hypothesis that immunosuppressed patients may be at increased risk for development of this disease.


Subject(s)
Brain Diseases/microbiology , Chromoblastomycosis/pathology , Cladosporium/isolation & purification , Liver Transplantation , Mycoses/pathology , Brain Diseases/epidemiology , Brain Diseases/pathology , Central Nervous System Diseases/epidemiology , Central Nervous System Diseases/microbiology , Central Nervous System Diseases/pathology , Chromoblastomycosis/epidemiology , Humans , Immunosuppression Therapy , Male , Middle Aged , Mycoses/epidemiology , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...