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.
Eur J Neurosci ; 41(7): 914-24, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25704649

ABSTRACT

Magnetoencephalography was recorded during a matching-to-sample plus cueing paradigm, in which participants judged the occurrence of changes in either categorical (CAT) or coordinate (COO) spatial relations. Previously, parietal and frontal lobes were identified as key areas in processing spatial relations and it was shown that each hemisphere was differently involved and modulated by the scope of the attention window (e.g. a large and small cue). In this study, Granger analysis highlighted the patterns of causality among involved brain areas--the direction of information transfer ran from the frontal to the visual cortex in the right hemisphere, whereas it ran in the opposite direction in the left side. Thus, the right frontal area seems to exert top-down influence, supporting the idea that, in this task, top-down signals are selectively related to the right side. Additionally, for CAT change preceded by a small cue, the right frontal gyrus was not involved in the information transfer, indicating a selective specialization of the left hemisphere for this condition. The present findings strengthen the conclusion of the presence of a remarkable hemispheric specialization for spatial relation processing and illustrate the complex interactions between the lateralized parts of the neural network. Moreover, they illustrate how focusing attention over large or small regions of the visual field engages these lateralized networks differently, particularly in the frontal regions of each hemisphere, consistent with the theory that spatial relation judgements require a fronto-parietal network in the left hemisphere for categorical relations and on the right hemisphere for coordinate spatial processing.


Subject(s)
Attention/physiology , Brain/physiology , Functional Laterality/physiology , Space Perception/physiology , Spatial Processing/physiology , Adult , Brain Mapping , Computer Simulation , Female , Humans , Information Theory , Magnetoencephalography , Male , Photic Stimulation , Signal Processing, Computer-Assisted , Young Adult
2.
Ital J Anat Embryol ; 110(2 Suppl 1): 127-33, 2005.
Article in English | MEDLINE | ID: mdl-16101030

ABSTRACT

Shedding of membrane vesicles is a vital phenomenon frequently observed in tumor cells and suggested to be involved in several aspects of tumor progression. Our previous studies have shown that human breast tumor cells rapidly shed membrane vesicles containing matrix metalloproteinases (MMPs). In this study we present that human umbilical vein endothelial cells (HUVEC) as well as different tumor cell lines (human ovarian cancer, CABA I and A2780, and hepatocarcinoma cell line, SK-Hep 1) shed vesicles in the extracellular medium. These vesicles carry MMPs and their inhibitors TIMPs. We conclude that tumor and endothelial cells shed MMP-containing vesicles and this may represent a mechanism for regulating focalized proteolytic activity and a way to interact with microenvironment during tumor angiogenesis.


Subject(s)
Cell Membrane/ultrastructure , Endothelial Cells/ultrastructure , Neoplasm Invasiveness/ultrastructure , Neoplasms/blood supply , Neovascularization, Pathologic/pathology , Secretory Vesicles/ultrastructure , Carcinoma/blood supply , Carcinoma/physiopathology , Carcinoma/ultrastructure , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/physiopathology , Carcinoma, Hepatocellular/ultrastructure , Cell Line, Tumor , Cell Membrane/physiology , Endothelial Cells/physiology , Exocytosis/physiology , Extracellular Space/metabolism , Female , Humans , Matrix Metalloproteinases/metabolism , Microscopy, Electron, Scanning , Neoplasm Invasiveness/physiopathology , Neoplasms/physiopathology , Neoplasms/ultrastructure , Neovascularization, Pathologic/physiopathology , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/physiopathology , Ovarian Neoplasms/ultrastructure , Secretory Vesicles/physiology , Tissue Inhibitor of Metalloproteinases/metabolism , Umbilical Veins
3.
J Exp Clin Cancer Res ; 23(1): 83-91, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15149155

ABSTRACT

Paclitaxel (PTX) is a potent anti-neoplastic agent that is highly effective in treating ovarian cancer. Nevertheless, the emergence of PTX resistance has limited the control of this disease. To gain insight into the molecular alterations accompanying drug resistance in ovarian cancer, we generated a new stable PTX-resistant ovarian carcinoma cell line. CABA I cells, which display an intrinsic PTX resistance (IC50 = 800 ng/ml), were subjected to continuous exposure to PTX. From the residual surviving cells, the highly PTX-resistant line CABA-PTX (IC50 = 256000 ng/ml) was generated and stably maintained in vitro. Analysis of beta-tubulin expression indicated that only the HM40 and Hbeta9 isotypes were expressed in both parental and resistant cells. No specific point mutations in the HM40 were detected in either cell line, but expression levels of this isotype were significantly reduced (40%) in CABA-PTX cells. Hbeta9 levels were unchanged. In those cells, PTX resistance was associated with cross-resistance to vinblastine but not to methotrexate or 5-fluorouracil. Verapamil treatment did not reverse the intrinsic drug resistance of parental cells, but partially modulated the sensitivity of CABA-PTX cells to PTX and induced total sensitivity to vinblastine. No changes in the cell surface expression of the drug efflux pumps MRP1, MRP2 and P-glycoprotein were observed. PTX influx, monitored using a fluorescent drug derivative, was significantly reduced and delayed in CABA-PTX cells as compared to the parental cells. Together, these findings suggest that more than one mechanism is involved in PTX resistance, making CABA-PTX cell line a potentially valuable in vitro tool to study multifactorial acquired drug resistance in ovarian cancer.


Subject(s)
Drug Resistance, Neoplasm , Ovarian Neoplasms/drug therapy , Paclitaxel/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Cell Division , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Flow Cytometry , Fluorouracil/pharmacology , Humans , Inhibitory Concentration 50 , Methotrexate/pharmacology , Mitochondrial Proteins/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Phenotype , Ribosomal Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Tetrazolium Salts/pharmacology , Time Factors , Tubulin/metabolism , Verapamil/pharmacology , Vinblastine/pharmacology
4.
Br J Cancer ; 86(7): 1180-7, 2002 Apr 08.
Article in English | MEDLINE | ID: mdl-11953869

ABSTRACT

Ovarian carcinomas represent a major form of gynaecological malignancies, whose treatment consists mainly of surgery and chemotherapy. Besides the difficulty of prognosis, therapy of ovarian carcinomas has reached scarce improvement, as a consequence of lack of efficacy and development of drug-resistance. The need of different biochemical and functional parameters has grown, in order to obtain a larger view on processes of biological and clinical significance. In this paper we report novel metabolic features detected in a series of different human ovary carcinoma lines, by (1)H NMR spectroscopy of intact cells and their extracts. Most importantly, a new ovarian adenocarcinoma line CABA I, showed strong signals in the spectral region between 3.5 and 4.0 p.p.m., assigned for the first time to the polyol sorbitol (39+/-11 nmol/10(6) cells). (13)C NMR analyses of these cells incubated with [1-(13)C]-D-glucose demonstrated labelled-sorbitol formation. The other ovarian carcinoma cell lines (OVCAR-3, IGROV 1, SK-OV-3 and OVCA432), showed, in the same spectral region, intense resonances from other metabolites: glutathione (up to 30 nmol/10(6) cells) and myo-inositol (up to 50 nmol/10(6) cells). Biochemical and biological functions are suggested for these compounds in human ovarian carcinoma cells, especially in relation to their possible role in cell detoxification mechanisms during tumour progression.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/analysis , Indicators and Reagents/pharmacokinetics , Magnetic Resonance Spectroscopy , Ovarian Neoplasms/pathology , Sorbitol/pharmacokinetics , Disease Progression , Female , Glutathione/metabolism , Humans , Hydrogen , Tumor Cells, Cultured
5.
Glycoconj J ; 17(3 -4): 261-8, 2000.
Article in English | MEDLINE | ID: mdl-11201799

ABSTRACT

This paper is the first report on the use of the electron microscopy autoradiography technique to detect metabolically tritium labeled sphingolipids in intact cells in culture. To label cell sphingolipids, human fibroblasts in culture were fed by a 24 hours pulse, repeated 5 times, of 3 x 10(-7) M [1-(3)H]sphingosine. [1-(3)H]sphingosine was efficently taken up by the cells and very rapidly used for the biosynthesis of complex sphingolipids, including neutral glycolipids, gangliosides, ceramide and sphingomyelin. The treatment with [1-(3)H]sphingosine did not induce any morphological alteration of cell structures, and well preserved cells, plasma membranes, and intracellular organelles could be observed by microscopy. Ultrathin sections from metabolic radiolabeled cells were coated with autoradiographic emulsion. One to four weeks of exposition resulted in pictures where the location of radioactive sphingolipids was evidenced by the characteristic appearance of silver grains as irregular coiled ribbons of metallic silver. Radioactive sphingolipids were found at the level of the plasma membranes, on the endoplasmic reticulum and inside of cytoplasmic vesicles. Thus, electron microscopy autoradiography is a very useful technique to study sphingolipid-enriched membrane domain organization and biosynthesis.


Subject(s)
Autoradiography/methods , Membrane Lipids/metabolism , Microscopy, Electron/methods , Sphingolipids/metabolism , Carbohydrate Sequence , Cells, Cultured , Fibroblasts , Humans , Isotope Labeling , Membrane Lipids/chemistry , Molecular Sequence Data , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/metabolism , Sphingolipids/chemistry , Sphingosine/chemistry , Sphingosine/metabolism , Tritium
6.
Clin Exp Metastasis ; 18(2): 163-70, 2000.
Article in English | MEDLINE | ID: mdl-11235992

ABSTRACT

The ability of a cell to modify the extracellular matrix is important in several pathophysiological alterations including tumorigenesis. Cell transformation is accompanied by changes in the surrounding stroma as a result of the action of specific proteases such as the urokinase plasminogen activator (uPA), which has been associated with invasive potential in many tumor types. In this study, we analyzed the release of vesicle-associated uPA by the aggressive prostatic carcinoma cell line PC3 and the implications of this release for the invasive behaviour of prostatic tumor cells. Zymography and Western blot analysis revealed the presence of vesicle-associated uPA in the high-molecular weight form. Vesicles adhered to and degraded both collagen IV and reconstituted basal membrane (Matrigel), and plasminogen enhanced the degradation in a dose-dependent manner. Addition of membrane vesicles shed by PC3 cells to cultures of the poorly invasive prostate cancer cell line LnCaP enhanced the adhesive and invasive capabilities of the latter, suggesting a mechanism involving substrate recognition and degradation. Together, these findings indicate that membrane vesicles can promote tumor invasion and point to the important role of vesicle-associated uPA in the extracellular compartment.


Subject(s)
Neoplasm Invasiveness/physiopathology , Prostatic Neoplasms/pathology , Urokinase-Type Plasminogen Activator/physiology , 3T3 Cells , Animals , Blotting, Western , Collagen/metabolism , Culture Media, Conditioned , Drug Combinations , Humans , Hydrolysis , Laminin , Male , Mice , Microscopy, Immunoelectron , Prostatic Neoplasms/metabolism , Proteoglycans , Tumor Cells, Cultured
7.
Clin Exp Metastasis ; 17(2): 131-40, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10411105

ABSTRACT

The in vitro release of matrix-degrading proteinases from breast cancer cells is associated in part with shed membrane vesicles. To determine whether shed vesicles might play a similar role in ovarian cancer cells, we analyzed the shedding phenomenon in vivo and in vitro as well as the enzymatic content of their vesicles. This is the first time that an immunoelectron microscopical analysis revealed membrane vesicles carrying tumor-associated antigen alpha-Folate Receptor (alpha-FR), circulating in biological fluids (ascites and serum) of an ovarian carcinoma patient. These vesicles were trapped in a fiber network with characteristic fibrin periodicity. An ovarian cancer cell line (CABA I) established from ascitic fluid cells of this patient, grew in Matrigel and formed tubular structures suggesting invasive capability. Immunofluorescence analysis demonstrated strong cytoplasmic staining of CABA I cells with anti-matrix metalloproteinase-9 (MMP-9) and anti-urokinase-type plasminogen activator (uPA) antibodies. CABA I cells shed membrane vesicles, which were morphologically similar to those identified in vivo, as determined by electron microscopy. Gelatin zymography of vesicles isolated both in vivo and in vitro revealed major gelatinolytic bands of the MMP family, identified as the zymogen and active forms of gelatinase B (MMP-9) and gelatinase A (MMP-2). By casein-plasminogen zymography we observed high-molecular weight (HMW)-uPA and plasmin bands. Incubation of purified vesicles from CABA I cells with Matrigel led to cleavage of Matrigel components. Taken together, our results point to a possible role of shed vesicles, both in vivo and in vitro, in proteolysis that mediates invasion and spread of ovarian epithelial carcinoma cells.


Subject(s)
Adenocarcinoma, Papillary/enzymology , Cell Membrane/ultrastructure , Metalloendopeptidases/metabolism , Ovarian Neoplasms/enzymology , Biomarkers, Tumor/analysis , Blotting, Western , Cell Membrane/enzymology , Female , Fluorescent Antibody Technique , Humans , In Vitro Techniques , Metalloendopeptidases/physiology , Microscopy, Electron , Middle Aged , Plasminogen Activators/metabolism , Tumor Cells, Cultured
8.
Int J Cancer ; 75(3): 418-31, 1998 Jan 30.
Article in English | MEDLINE | ID: mdl-9455804

ABSTRACT

Bombesin is a potent inducer of signal trasduction pathways involved in the proliferation and invasion of androgen-insensitive prostatic tumor cells. This study examines the bombesin-mediated modulation of pericellular proteolysis, monitoring cell capability to migrate and invade basement membranes, using a chemo-invasion assay and analyzing protease production. The results suggest that bombesin could modulate the invasive potential of prostatic cell lines regulating secretion and cell-surface uptake of uPA and MMP-9 activation. In fact, in PC3 and DU145 cells but not in LNCaP cells, urokinase-type plasminogen activator (uPA) and plasminogen activator inhibitor-1 (PAI-1) are induced by bombesin treatment. Bombesin also stimulates cell proliferation and this effect can be inhibited blocking uPA by antibodies and/or uPA inhibitor p-aminobenzamidine. Moreover, HMW-uPA induces cell proliferation in LNCaP cells, which do not produce uPA in the basal conditions, while PC3 and DU145 cell growth is supported by autocrine production of uPA. The increment of uPA activity on the external plasma membrane causes an increased pericellular plasmin activation. This effect is inhibited by antibodies against uPA and by p-aminobenzamidine. Similarly to EGF, bombesin stimulates secretion and activation of MMP-9 and TIMP-1 production. MMP-9 activation can be also obtained by HMW-uPA treatment, suggesting that plasma-membrane-bound uPA can start a proteolytic cascade involving MMP-9. Therefore, in in vitro assays, bombesin is able to modulate pericellular proteolysis and cell proliferation, differently distributing and activating proteolytic activities. This effect can be related to the "non-random" degradation of the extracellular matrix in which membrane uPA-uPAreceptor complexes could start bombesin-induced directional protein degradation during metastatic spread.


Subject(s)
Antineoplastic Agents/pharmacology , Bombesin/pharmacology , Peptide Hydrolases/drug effects , Peptide Hydrolases/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Antibodies/pharmacology , Benzamidines/pharmacology , Cell Division/drug effects , Cell Membrane/enzymology , Collagen , Collagenases/biosynthesis , Culture Media, Conditioned , Drug Combinations , Enzyme Activation , Enzyme Induction , Epidermal Growth Factor/pharmacology , Extracellular Matrix/metabolism , Fibrinolysin/metabolism , Gastrin-Releasing Peptide/pharmacology , Humans , Laminin , Male , Matrix Metalloproteinase 9 , Neoplasm Invasiveness , Neoplasm Metastasis , Peptide Hydrolases/biosynthesis , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/metabolism , Prostatic Neoplasms/pathology , Proteoglycans , Serine Proteinase Inhibitors/pharmacology , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/biosynthesis , Urokinase-Type Plasminogen Activator/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...