Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Endocr Relat Cancer ; 14(4): 1029-38, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18045954

ABSTRACT

Breast cancer cells are usually sensitive to several chemotherapeutic regimens, but they can develop chemoresistance after prolonged exposure to cytotoxic drugs, acquiring a more aggressive phenotype. Drug resistance might involve the multi-drug resistance (MDR) 1 gene, encoding a transmembrane glycoprotein p-170 (P-gp), which antagonizes intracellular accumulation of cytotoxic agents, such as doxorubicin. We previously demonstrated that type 2 cyclooxygenase (COX-2) inhibitors can reverse the chemoresistance phenotype of a medullary thyroid carcinoma cell line by inhibiting P-gp expression and function. The aim of our study was to investigate the role of COX-2 inhibitors in modulating chemoresistance in a human breast cancer cell line, MCF7. MCF7 cells, expressing COX-2 but not MDR1, were treated with increasing doses of doxorubicin, and they became chemoresistant after 10 days of treatment, in association with MDR1 expression induction. This effect was reversed by doxorubicin withdrawal and prevented by co-incubation with N-[2-(cyclohexyloxy)4-nitrophenyl]-methanesulfonamide (NS-398), a selective COX-2 inhibitor. Treatment with NS-398 alone did not influence cell viability of a resistant MCF7 cell clone (rMCF7), but sensitized rMCF7 cells to the cytotoxic effects of doxorubicin. Moreover, treatment with NS-398 significantly reduced MDR1 expression in rMCF7 cells. Doxorubicin-induced membrane P-gp expression and function was also greatly impaired. Our data therefore support the hypothesis that COX-2 inhibitors can prevent or reduce the development of the chemoresistance phenotype in breast cancer cells by inhibiting P-gp expression and function.


Subject(s)
Breast Neoplasms/genetics , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/genetics , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic/drug effects , Glycoproteins/genetics , Nitrobenzenes/pharmacology , Sulfonamides/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclooxygenase Inhibitors/pharmacology , Doxorubicin/pharmacokinetics , Doxorubicin/toxicity , Female , Humans , Phenotype , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction
2.
J Cell Physiol ; 212(2): 293-7, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17443684

ABSTRACT

The aminoacyl t-RNA synthetase interacting multifunctional protein (AIMP1) is the precursor of the multifunctional inflammatory cytokine endothelial monocyte-activating polypeptide II (EMAP II). We previously demonstrated that AIMP1 secretion by pituitary adenomas is inversely correlated with tumor diameter and with RARS expression, suggesting that a high amount of RARS associated with AIMP1 might prevent the secretion of the latter cytokine. In this study, we investigated the role of RARS in modulating the secretion of AIMP1 in HeLa and MCF7 cell lines and investigated the possible role of the multicatalytic protease in the cleavage of AIMP1 to generate EMAP II. Our data show that RARS over-expression impairs AIMP1 secretion by both HeLa and MCF7 cells. Moreover, proteasome inhibition impairs AIMP1 cleavage to produce EMAP II. These data indicate that RARS over-expression associates with a reduced AIMP1 secretion and that the multicatalytic protease is involved in the generation of the mature cytokine, EMAP II.


Subject(s)
Arginine-tRNA Ligase/metabolism , Breast Neoplasms/metabolism , Cytokines/metabolism , Neoplasm Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , RNA-Binding Proteins/metabolism , Uterine Cervical Neoplasms/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Arginine-tRNA Ligase/genetics , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cysteine Proteinase Inhibitors/pharmacology , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Proteasome Inhibitors , Protein Processing, Post-Translational/drug effects , Transfection , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/genetics
3.
Endocrinology ; 147(7): 3530-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16601140

ABSTRACT

Somatostatin (SRIH) inhibits cell proliferation by interacting with five distinct SRIH receptor subtypes (SSTRs) activating several pathways in many tissues. We previously demonstrated that SRIH, by activating Src homology-2-containing protein, inhibits cell proliferation of the human medullary thyroid carcinoma cell line, TT, which expresses all SSTRs. However, the effects of SRIH on cell cycle proteins have not been investigated so far. We therefore evaluated the effects of SRIH and a selective SSTR2 agonist on cell cycle protein expression, mainly focusing on cyclin D1 and its associated kinases. Our data show that SRIH and the selective SSTR2 agonist, BIM-23120, reduce cell proliferation and DNA synthesis as well as induce a delay of the cell cycle in G(2)/M phase. Moreover, treatment with both SRIH and BIM-23120 decreases cyclin D1 levels, with a parallel increase in phosphocyclin D1 levels, suggesting protein degradation. Moreover, our data show an increase in glycogen synthase kinase-3beta activity, which triggers phosphorylation-dependent cyclin D1 degradation. Indeed, we observed a reduction in cyclin D1 protein half-life under treatment with SRIH or the SSTR2 selective agonist. A reduction in cdk4 protein levels is also observed with a parallel reduction in Rb phosphorylation levels at Ser-780. Our data indicate that the subtype 2 receptor-mediated antiproliferative effect of SRIH on TT cell proliferation may be exerted through a decrease in cyclin D1 levels.


Subject(s)
Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Thyroid Neoplasms/metabolism , Cell Division , Cell Line, Tumor , Cell Proliferation , Cell Survival , DNA/metabolism , G2 Phase , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , In Vitro Techniques , Retinoblastoma Protein/metabolism
4.
J Clin Endocrinol Metab ; 91(6): 2218-24, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16569735

ABSTRACT

CONTEXT: Medullary thyroid carcinoma (MTC) is a rare tumor originating from thyroid parafollicular C cells. We previously demonstrated that somatostatin (SRIH) reduces cell growth in the human MTC cell line, TT, which expresses all SRIH receptor (SSTR) subtypes and responds differently to selective SSTR agonists. OBJECTIVE: To clarify the possible effects of SRIH analogs on hormone secretion and proliferation in MTC primary cultures, we evaluated SSTR expression and assessed the in vitro effects on calcitonin (CT) and chromogranin A secretion as well as cell viability of SRIH analogs interacting with SSTR1, SSTR2, and SSTR5. DESIGN: Thirty-five patients affected by MTC were recruited from 2003 to 2005. After total thyroidectomy, the samples were examined for CT, chromogranin A, and SSTR expression by RT-PCR. Primary cultures were developed and tested with SRIH analogs interacting with SSTR1, SSTR2, and SSTR5. RESULTS: We selected 18 MTC tumor samples, expressing SSTR1, SSTR2, and SSTR5. Two different groups were identified according to CT secretion inhibition by the clinically available SRIH analog, lanreotide. In the responder group, CT secretion was reduced by compounds interacting with SSTR1, SSTR2, and SSTR5, whereas cell viability was not affected. On the other hand, in the nonresponder group, CT secretion was reduced by the SSTR1 selective agonist, whereas cell viability was inhibited by SSTR2 selective agonists. CONCLUSIONS: Our data suggest that SRIH analogs might be useful in medical therapy of MTC because they could have antiproliferative effects despite the lack of antisecretory activity and vice versa.


Subject(s)
Calcitonin/metabolism , Carcinoma, Medullary/drug therapy , Chromogranins/metabolism , Receptors, Somatostatin/agonists , Thyroid Neoplasms/drug therapy , Adult , Aged , Carcinoma, Medullary/metabolism , Carcinoma, Medullary/pathology , Cell Line, Tumor , Cell Survival/drug effects , Child , Chromogranin A , Female , Humans , Male , Middle Aged , Receptors, Somatostatin/analysis , Receptors, Somatostatin/classification , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
5.
J Clin Endocrinol Metab ; 90(10): 5754-60, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16091477

ABSTRACT

OBJECTIVE: Medullary thyroid carcinoma (MTC) is a highly chemoresistant malignant neoplasia deriving from parafollicular C cells. Chemotherapy failure has been ascribed, at least in part, to the overexpression by MTC of the multidrug resistance 1 (MDR1) gene, encoding a transmembrane glycoprotein [permeability glycoprotein (P-gp)] that antagonizes intracellular accumulation of cytotoxic agents. P-gp expression and function in a rat model have been demonstrated to depend on cyclooxygenase (COX)-2 isoform levels, which are found elevated in many human cancers. The aim of our study was to investigate the role of the COX-2 pathway in modulating chemoresistance. DESIGN AND RESULTS: We investigated P-gp and COX-2 expression and then evaluated the sensitizing effects of COX-2 inhibitors on the cytotoxic effects of doxorubicin in the presence or in the absence of prostaglandin E2 in primary cultures and in a human MTC cell line, TT. Moreover, P-gp function has been studied. Our data show that TT cells express both MDR1 and COX-2 and that rofecoxib, a selective COX-2 inhibitor, sensitizes TT cells to the cytotoxic effects of doxorubicin, reducing P-gp expression and function. CONCLUSIONS: Our data suggest that these effects are mediated by a mechanism not involving the generation of prostaglandin E2, possibly implicating the synthesis of other COX-2 products.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Carcinoma, Medullary/drug therapy , Carcinoma, Medullary/metabolism , Cyclooxygenase Inhibitors/pharmacology , Drug Resistance, Neoplasm , Prostaglandin-Endoperoxide Synthases , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/pharmacology , Blotting, Western , Calcium Channel Blockers/pharmacology , Cell Count , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Dinoprostone/pharmacology , Doxorubicin/pharmacology , Humans , Lactones/pharmacology , Membrane Proteins , Permeability , Phenotype , RNA, Neoplasm/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Sulfones/pharmacology , Verapamil/pharmacology
6.
Endocrinology ; 146(6): 2692-8, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15746253

ABSTRACT

Medullary thyroid carcinoma (MTC) is a rare tumor originating from thyroid parafollicular C cells, where, in the inherited form, constitutive activation of the RET protooncogene is responsible for unrestrained cell proliferation. We previously demonstrated that somatostatin (SRIF) reduces cell growth in the human MTC cell line TT, which expresses all SRIF receptor (SSTR) subtypes and responds differently to selective SSTR agonists. The antiproliferative mechanism of SRIF and its analogs in MTC is still unclear. Src homology-2-containing protein tyrosine phosphatase-1 (SHP-1), a cytoplasmic protein tyrosine phosphatase (PTP), is activated by somatotropin release-inhibiting factor and reduces mutated RET autophosphorylation in a heterologous system. In this study, we explore the role of PTP activation, in particular of SHP-1, in TT cells, where RET is constitutively activated. In TT cells, SRIF stimulated the PTP activity of SHP-1, which was associated with proliferation inhibition and with reduction in the MAPK pathway activation. Blockade of PTP activity with sodium orthovanadate induced cell proliferation and MAPK phosphorylation and blunted the inhibitory effects of SRIF. Moreover, SHP-1 associates with SSTR2 depending on its activation. By using a MAPK kinase inhibitor, we demonstrated that TT cell growth depends on MAPK pathway activation. Furthermore, in TT cells overexpressing SHP-1, cell proliferation and MAPK signaling were strongly down-regulated, whereas in TT cells transfected with a dominant negative form of SHP-1, cell proliferation and MAPK signaling were markedly induced. Our data demonstrate that SRIF inhibitory effects on TT cell proliferation are mediated, at least in part, by SHP-1, which acts through a MAPK-dependent mechanism.


Subject(s)
Carcinoma, Medullary/metabolism , Carcinoma, Medullary/pathology , Protein Tyrosine Phosphatases/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Cell Division/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System/physiology , Phosphorylation , Protein Phosphatase 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/genetics , Receptors, Somatostatin/metabolism
7.
J Cell Physiol ; 204(1): 280-5, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15648093

ABSTRACT

Micro RNAs (miRs) are small noncoding RNAs, functioning as antisense regulators of other RNAs. miR-15a and miR-16-1 genes are located at chromosome 13q14, a region which is frequently deleted in pituitary tumors. An inverse correlation has been shown in B cell chronic lymphocytic leukemia (B-CLL) between miR-15a and miR-16-1 expression and the expression levels of arginyl-tRNA synthetase (RARS), an enzyme which associates with the cofactor p43 in the aminoacyl-tRNA synthetase complex. When secreted, p43 regulates local inflammatory response and macrophage chemotaxis, and seems to have anti-neoplastic properties in mice. We explored miR-15a and miR-16-1 expression in 10 GH-secreting and in 10 PRL-secreting pituitary macroadenomas by Northern blot, and investigated the possible correlation with in vivo and in vitro characteristics. We found that miR-15a and miR-16-1 are expressed at lower levels in pituitary adenomas as compared to normal pituitary tissue. Moreover, their expression inversely correlates with tumor diameter and with RARS expression (P < 0.05), but directly correlates with p43 secretion (P < 0.02). Therefore, miR15 and miR16 down-regulation in pituitary adenomas correlates with a greater tumor diameter and a lower p43 secretion, suggesting that these genes may, at least in part, influence tumor growth.


Subject(s)
Adenoma/genetics , Gene Expression Regulation, Neoplastic/physiology , MicroRNAs/physiology , Pituitary Gland/physiology , Pituitary Neoplasms/genetics , Adenoma/physiopathology , Adolescent , Adult , Aged , Antigens, Neoplasm , Arginine-tRNA Ligase/genetics , Down-Regulation , Female , Gene Deletion , Humans , Male , Middle Aged , Mitochondrial Proteins , Peptide Elongation Factor Tu/metabolism , Pituitary Neoplasms/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...