Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Biochem ; 65(4): 479-91, 1997 Jun 15.
Article in English | MEDLINE | ID: mdl-9178098

ABSTRACT

We investigated whether the folate receptor alpha-isoform (FR alpha), which is overexpressed on ovarian carcinoma cells, is functionally active in internalizing the physiological form et folate, 5-methyl tetrahydrofolate (THF). Six ovarian tumor cell lines, expressing different levels of FR alpha (COR > > OVCAR3 > IGROV1 > OVCAR4 > SKOV3 > OVCAR5), were maintained in folate-depleted medium and internalization of 10 nM evaluated as acid-resistant radioactivity at 0 degree and 37 degrees C. The amount of 5-methyl[1H]THF present in this fraction was not strictly related to the number of membrane receptors, since even cell lines with low FR alpha expression, e.g., OVCAR4, showed efficient internalization. Time-course studies indicated that, whereas no uptake was detected at 0 degree C, at 37 degrees C the internalized fraction showed a slow and constant increase, until 4 h. At this time the internalized radioactivity represented < 50% of the total bound in COR, OVCAR3 and IGROV1 cells, whereas the other cell lines tested internalized fourfold more folate than their surface binding capacity. The incubation in the presence of a concentration (50 nM) of 5-methyl[3H]THF, which best ensures receptors saturation on cells with highest FR levels (COR and OVCAR3), had slight effect on surface binding of all the tested cell lines, including IGROV1 and SKOV3. In contrast, the increase of the uptake was more pronounced, particularly in SKOV3 cells. These results, together with the accumulation curves of folic acid (FA) and 5-methylTHF at 37 degrees C, suggested the presence of a molecule on ovarian carcinoma cells with high affinity for reduced folates, possibly a reduced folate carrier (RFC). Measurement of radioactivity present in the supernatant of IGROV1 and SKOV3 cells, subjected to hypotonic lysis and cell fractionation, further indicated that 5-methyl[3H]THF was translocated to the cytosol and, despite differences in membrane levels of FR alpha expression this internalized fraction was similar in both cell lines. Inhibition experiments to selectively block FR alpha or RFC activity showed a differential sensitivity of the two pathways depending on the cell line examined. Internalization was more consistently inhibited on IGROV1 than on SKOV3 cells by treatments that disrupt FR alpha activity, e.g., incubation with excess FA and phosphatidylinositol specific phospholipase C, whereas Probenecid, which preferentially inhibits the carrier-mediated pathway, showed a strong inhibitory effect on both cell lines. These findings suggest that the internalization of 5-methylTHF in these tumor cells depends not only on the level of overexpressed FR alpha, but another transport route, with features characteristic for RFC, is functional and participates in folate uptake.


Subject(s)
Folic Acid/metabolism , Ovarian Neoplasms/metabolism , Receptors, Cell Surface , Biological Transport , Carrier Proteins/metabolism , Cell Membrane/metabolism , Cytosol/metabolism , Female , Folate Receptors, GPI-Anchored , Humans , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoinositide Phospholipase C , Probenecid/pharmacology , Tetrahydrofolates/metabolism , Tumor Cells, Cultured , Type C Phospholipases/metabolism , Uricosuric Agents/pharmacology
2.
Br J Cancer ; 76(1): 77-82, 1997.
Article in English | MEDLINE | ID: mdl-9218736

ABSTRACT

It has been suggested that sensitivity of ovarian carcinomas to cisplatin is in part related to an endogenous folate deficiency. In this work, we investigated whether overexpression of the folate-binding protein (FBP), a receptor involved in folate transport, might be associated with cisplatin sensitivity. The results obtained on a panel of ten ovarian carcinoma cell lines that overexpress different levels of the FBP showed a statistically significant relationship between FBP overexpression and cisplatin responsiveness, with the most sensitive cell lines expressing higher FBP levels on their membrane than the less sensitive ones. The relationship was observed both in cells growing in standard medium-containing high-folate concentrations (2.3 microM) and in cells adapted to growth in low-folate (20 nM) medium. Analysis of two cisplatin-resistant cell lines derived from the cisplatin-sensitive IGROV1 ovarian carcinoma cell line indicated that resistance was associated with a significant decrease in FBP expression. However, the receptor does not appear to be directly responsible for drug sensitivity per se as different cell lines transfected with FBP cDNA did not become more sensitive to the drug. Together, the data suggest the possible predictive value of FBP in ovarian carcinoma, as higher levels of expression can be indirectly but significantly associated with increased drug sensitivity.


Subject(s)
Carrier Proteins/metabolism , Cisplatin/pharmacology , Ovarian Neoplasms/metabolism , Receptors, Cell Surface/metabolism , Carrier Proteins/genetics , Drug Resistance, Neoplasm , Female , Fluorescent Antibody Technique, Indirect , Folate Receptors, GPI-Anchored , Humans , Ovarian Neoplasms/drug therapy , Transfection , Tumor Cells, Cultured
3.
Cancer Res ; 56(4): 689-93, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8630996

ABSTRACT

The p53 protein is a multifunctional transcriptional regulator involved in cellular response to DNA damage and has been implicated as a putative determinant of sensitivity of tumor cells to cytotoxic agents. Since the p53 gene becomes inactivated in over one-half of advanced ovarian carcinoma, in this study we have examined the relationships between p53 gene alterations, p53 immunoreactivity, and response to cisplatin-based chemotherapy in ovarian cancer patients. All patients had advanced (FIGO stage III or IV) ovarian carcinoma and, with one exception, were untreated at the time of collection of tumor specimens. After initial debulking surgery, patients received high-dose cisplatin therapy. Tumor samples were analyzed for p53 gene mutations and for p53 protein accumulation, and the findings were correlated with tumor responsiveness. Of the 33 tumors examined, p53 gene mutations were found in 20 cases, including 15 missense mutations, 2 deletions, 2 nonsense mutations, and a base substitution at splice site. Twenty tumors showed positive immunostaining for p53. Only missense mutations were associated with positive immunostaining. In addition, p53 overexpression was detected in five tumors in the absence of mutations. Most (12 of 14) of the missense mutations associated with p53 protein stabilization were found refractory to therapy, as well as tumors overexpressing wild-type p53 (4 of 5). A significant correlation has been found between p53 accumulation, type of mutation (i.e., missense mutations), and pathological response to cisplatin-based therapy. In conclusion, the present results are consistent with a role of p53 as a determinant of chemosensitivity of ovarian carcinoma.


Subject(s)
Cisplatin/therapeutic use , Genes, p53 , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Point Mutation , Sequence Deletion , Tumor Suppressor Protein p53/biosynthesis , Alleles , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Codon , Exons , Female , Humans , Immunohistochemistry , Immunophenotyping , Introns , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/pathology , Ovarian Neoplasms/surgery
4.
Int J Cancer ; 63(3): 395-401, 1995 Nov 03.
Article in English | MEDLINE | ID: mdl-7591238

ABSTRACT

Ovarian-carcinoma cell lines (OVCAR3, IGROVI, OVCA432, SW626 and SKOV3), grown in standard medium containing supra-physiological (2.3 microM) folate concentration, display different levels of reactivity with the anti-folate-binding-protein (FBP) monoclonal antibody MOv18, which recognizes the alpha-isoform of the protein. Gel-filtration and absorption experiments indicated that on IGROVI cells this molecule accounts for all folic-acid binding at nanomolar concentrations. The aim of the study was to investigate the effect of extracellular folate levels on cells adapted to grow in medium containing physiological folate concentration (20 nM). By the ternary complex assay, all cell lines showed a marked depletion of intracellular reduced folates, compared with those in standard folate medium. The monitoring of FBP by MOv18 showed on IGROVI cells a transient up-regulation of the protein, whereas on the other cell lines, except SKOV3, no changes were detected. These data suggest that in these cells further over-expression of the molecule cannot generally be induced by lowering the extracellular folate concentration. On SKOV3, Scatchard analysis of 125I-MOv18 binding, as well as the evaluation of total folate binding capacity, showed a 2- to 3-fold stable increase of FBP expression after long-term growth in low-folate medium. Reverse transcriptase-polymerase chain reaction (RT-PCR) analysis indicated in these cells a 1.5-fold increase in alpha-FBP mRNA. SKOV3 cells, maintained in vitro in medium containing supraphysiological and physiological (i.e., low-folate) concentrations were injected into nude mice. Weight differences, though not statistically significant, were observed in favour of low-folate-derived tumors. Immunohistochemical and immunochemical analysis of the tumor samples showed that in SKOV3 cells the receptor modulation can also be induced by restoring the physiological folate concentration in vivo.


Subject(s)
Carrier Proteins/analysis , Folic Acid/pharmacology , Ovarian Neoplasms/pathology , Receptors, Cell Surface , Animals , Base Sequence , Carrier Proteins/genetics , Cell Division/drug effects , Female , Folate Receptors, GPI-Anchored , Folic Acid/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Ovarian Neoplasms/chemistry , RNA, Messenger/analysis , Tumor Cells, Cultured
5.
Hum Hered ; 45(2): 75-9, 1995.
Article in English | MEDLINE | ID: mdl-7750978

ABSTRACT

The chromosomal localisation of the human gene coding for peroxisomal 3-oxoacyl-CoA thiolase (ACAA) was determined by human-hamster somatic cell hybrids and fluorescence in situ hybridisation, using cDNA and genomic probes, respectively. The results allowed an exclusion of the previously suggested presence of a second site for ACAA on chromosome 11 and an assignment of the gene to a single chromosome band (3p22).


Subject(s)
Acetyl-CoA C-Acyltransferase/genetics , Chromosome Mapping/methods , Chromosomes, Human, Pair 3 , Microbodies/enzymology , Animals , Cricetinae , DNA Probes , Humans , Hybrid Cells , In Situ Hybridization, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL
...