Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Front Biosci (Landmark Ed) ; 18(1): 133-44, 2013 01 01.
Article in English | MEDLINE | ID: mdl-23276914

ABSTRACT

Breast cancer is a major cause of cancer death in women in the world. Triple-negative breast cancers, which accounts for 10-20% of all mammary tumours, are characterised by an aggressive phenotype, are often found in younger women and have been associated with poor prognosis. Obesity increases the risk for triple-negative breast cancer occurrence. Because triple-negative breast cancer patients are unresponsive to current targeted therapies and other treatment options are only partially effective, new pharmacological approaches are warranted. The obesity-linked adipokine, leptin, is a well known mitogen/survival factor in breast cancer cells and several studies have addressed the role of leptin in breast cancer pathogenesis and progression. Surprisingly, recent in vitro studies have shown that leptin enhances the anti-proliferative effects of cAMP elevation in triple-negative breast cancer cells by apoptosis induction. In the current review, we discuss on the role of cAMP as a growth suppressor and of leptin as a growth promoting factor in breast cancer cells and we will focus on the molecular pathways involved in the antiproliferative interaction between leptin and cAMP elevation. The rationale for the possible development of a simple, cheap and innovative approach for therapeutic intervention in triple-negative breast cancer, based on the use of cAMP elevating drugs at lower and tolerable doses, will be also discussed.


Subject(s)
Breast Neoplasms/drug therapy , Cyclic AMP/physiology , Leptin/physiology , Signal Transduction/drug effects , Breast Neoplasms/physiopathology , Cell Line, Tumor , Cyclic AMP/agonists , Cyclic AMP/therapeutic use , Drug Resistance, Neoplasm/drug effects , Female , Humans , Leptin/therapeutic use , STAT3 Transcription Factor/metabolism
2.
J Cell Physiol ; 228(1): 198-206, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22674530

ABSTRACT

Osteosarcoma is the most common malignant primary bone tumor in children and adolescents. The clinical outcome for osteosarcoma remains discouraging despite aggressive surgery and intensive radiotherapy and chemotherapy regimens. Thus, novel therapeutic approaches are needed. Previously, we have shown that inorganic phosphate (Pi) inhibits proliferation and aggressiveness of human osteosarcoma U2OS cells identifying adenylate cyclase, beta3 integrin, Rap1, ERK1/2 as proteins whose expression and function are relevantly affected in response to Pi. In this study, we investigated whether Pi could affect chemosensitivity of osteosarcoma cells and the underlying molecular mechanisms. Here, we report that Pi inhibits proliferation of p53-wild type U2OS cells (and not of p53-null Saos and p53-mutant MG63 cells) by slowing-down cell cycle progression, without apoptosis occurrence. Interestingly, we found that Pi strongly enhances doxorubicin-induced cytotoxicity in U2OS, and not in Saos and MG63 cells, by apoptosis induction, as revealed by a marked increase of sub-G1 population, Bcl-2 downregulation, caspase-3 activation, and PARP cleavage. Remarkably, Pi/doxorubicin combination-induced cytotoxicity was accompanied by an increase of p53 protein levels and of p53 target genes mdm2, p21 and Bax, and was significantly reduced by the p53 inhibitor pifithrine-alpha. Moreover, the doxorubicin-induced cytotoxicity was associated with ERK1/2 pathway inhibition in response to Pi. Altogether, our data enforce the evidence of Pi as a novel signaling molecule capable of inhibiting ERK pathway and inducing sensitization to doxorubicin of osteosarcoma cells by p53-dependent apoptosis, implying that targeting Pi levels might represent a rational strategy for improving osteosarcoma therapy.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Bone Neoplasms/drug therapy , Doxorubicin/therapeutic use , Osteosarcoma/drug therapy , Phosphates/therapeutic use , Tumor Suppressor Protein p53/metabolism , Antibiotics, Antineoplastic/administration & dosage , Bone Neoplasms/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Doxorubicin/administration & dosage , Drug Therapy, Combination , Gene Expression Regulation, Neoplastic/drug effects , Humans , Osteosarcoma/metabolism , Phosphates/administration & dosage , RNA Interference , RNA, Small Interfering , Tumor Suppressor Protein p53/genetics
3.
Front Biosci (Elite Ed) ; 3(4): 1249-58, 2011 06 01.
Article in English | MEDLINE | ID: mdl-21622131

ABSTRACT

Osteosarcoma is the most common malignant primary bone tumor in children and adolescents and is characterized by a high metastatic potential. Its clinical outcome remains discouraging despite aggressive treatments. Thus, novel therapeutic approaches are needed. Recent results indicate that inorganic phosphate (Pi) is capable of affecting specific signal transduction pathways and of acting as an active regulator of cell behaviour. Previously, we found that Pi inhibits proliferation of human osteosarcoma U2OS cells via an adenylate cyclase/cAMP mediated mechanism. Here, we report that upon Pi treatment, U2OS cells become extremely hard to dislodge with trypsin. The lack of sensitivity to the trypsin action was paralleled by relevant changes in integrin subunits expression and accompanied by an increase of cell adhesion in cell-matrix adhesion assays. Interestingly, exposure of U2OS cells to Pi results also in a strong activation and protein level up-regulation of Rap1 small GTPase and in an early increase followed by a sustained inhibition of Erk1/2 phosphorylation. Importantly, the Pi-induced increase of cell adhesion was enforced by a cAMP analogue which specifically activated Epac/Rap1 and insensitive to PKA and MEK1/2 inhibitors. Our results enforce the evidences of inorganic phosphate as a signalling molecule, identify beta3 integrin, Rap1, ERK1/2 as proteins whose expression and function are relevantly affected by Pi in osteosarcoma U2OS cells The clinical significance and potential therapeutic applications by our data will be discussed.


Subject(s)
Osteosarcoma/pathology , Phosphates/pharmacology , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Humans , Osteosarcoma/metabolism
4.
J Cell Physiol ; 225(3): 801-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20589829

ABSTRACT

Previously, we have shown that leptin potentiates the antiproliferative action of cAMP elevating agents in breast cancer cells and that the protein kinase A (PKA) inhibitor KT-5720 prevented the antiproliferative effects induced by the leptin plus cAMP elevation. The present experiments were designed to gain a better understanding about the PKA role in the antitumor interaction between leptin and cAMP elevating agents and on the underlying signaling pathways. Here we show that exposure of MDA-MB-231 breast cancer cells to leptin resulted in a strong phosphorylation of both ERK1/2 and STAT3. Interestingly, intracellular cAMP elevation upon forskolin pretreatment completely abrogated both ERK1/2 and STAT3 phosphorylation in response to leptin and was accompanied by a consistent CREB phosphorylation. Notably, leptin plus forskolin cotreatments resulted in a strong decrease of both PKA regulatory RIα and catalytic subunits protein levels. Importantly, pretreatment with the PKA inhibitor KT-5720 blocked the forskolin-induced CREB phosphorylation and prevented both the inhibition by forskolin of leptin-induced ERK1/2 and STAT3 phosphorylation and the PKA subunits down-regulation induced by the combination of leptin and forskolin. Altogether, our results indicate that leptin-dependent signaling pathways are influenced by cAMP elevation and identify PKA as relevantly involved in the pharmacological antitumor interaction between leptin and cAMP elevating drugs in MDA-MB-231 cells. We propose a molecular model by which PKA confers its effects. Potential therapeutic applications by our data will be discussed.


Subject(s)
Breast Neoplasms/enzymology , Cell Proliferation , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Leptin/metabolism , Signal Transduction , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Carbazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Colforsin/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Down-Regulation , Enzyme Activators/pharmacology , Female , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Recombinant Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Time Factors , Up-Regulation
5.
Cancer Biol Ther ; 8(12): 1183-90, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19662684

ABSTRACT

Elevation of cAMP inhibits the proliferation and expression of transformed phenotype in several cell types, including breast cancer cells. Leptin has been shown to act as a mitogen/survival factor in many types of cancer cells. In the present work, we have studied the impact of cAMP elevation on leptin-induced proliferation of breast cancer cells. Here we report that treatment of estrogen receptor negative human breast cancer cell line MDA-MB-231 with leptin or cAMP elevating agents has positive and negative effects on cell proliferation, respectively. Surprisingly, we find that leptin strongly potentiates the anti-proliferative action of cAMP elevating agents, by concurring to cell cycle arrest at G1 phase and inducing apoptosis. Pretreatment with the PKA inhibitor KT-5720 completely prevented the anti-proliferative effects induced by the combination between leptin and cAMP elevating agents. The above anti-proliferative effects were paralleled by the decrease of cyclin D1 and A and by the increase of inhibitor p27kip1 cell cycle regulating protein levels. In these conditions we found also a strong decrease of anti-apopotic Bcl2 protein levels. Altogether, our data extend the evidence of adenylate cyclase/cAMP/PKA as a growth suppressor system and of leptin as a growth promoting factor in breast cancer cells. Remarkably, our results suggest that when cAMP levels are increased, leptin drives cells towards apoptosis, and that targeting both cAMP levels and leptin signalling might represent a simple novel way for therapeutic intervention in breast cancer.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cyclic AMP/metabolism , Leptin/pharmacology , 1-Methyl-3-isobutylxanthine/antagonists & inhibitors , 1-Methyl-3-isobutylxanthine/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/antagonists & inhibitors , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Breast Neoplasms/pathology , Carbazoles/pharmacology , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Colforsin/antagonists & inhibitors , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , G1 Phase/drug effects , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrroles/pharmacology , Signal Transduction
6.
Expert Opin Ther Targets ; 13(1): 83-92, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19063708

ABSTRACT

BACKGROUND: cAMP is a second messenger that plays a role in intracellular signal transduction of various stimuli. a major function of cAMP in eukaryotes is activation of cAMP-dependent protein kinase (PKA). PKA is the best understood member of the serine-threonine protein kinase superfamily, and is involved in the control of a variety of cellular processes. since it has been implicated in the initiation and progression of many tumors, PKA has been suggested as a novel molecular target for cancer therapy. OBJECTIVE/METHODS: Here, after describing some features of cAMP/PKA signaling that are relevant to cancer biology, we review targeting of PKA in cancer therapy, also discussing PKA as a biomarker for cancer detection and monitoring of therapy. RESULTS/CONCLUSIONS: PKA is an increasingly relevant biological target in the therapy and management of cancer.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Cyclic AMP/metabolism , Humans , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...