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










Publication year range
1.
J Cell Physiol ; 235(4): 3741-3752, 2020 04.
Article in English | MEDLINE | ID: mdl-31602671

ABSTRACT

Osteosarcoma (OS) is a very aggressive metastatic pediatric and adolescent tumor. Due to its recurrent development of chemotherapy resistance, clinical outcome for OS patients remains poor. Therefore, discovering more effective anticancer agents is needed. Chlorogenic acid (CGA) is a phenolic compound contained in plant-related products that modulates many cellular functions and inhibits cell proliferation in several cancer types. However, few evidence is available in OS. Here, we investigate the effects of CGA in U2OS, Saos-2, and MG-63 OS cells. By multiple approaches, we demonstrate that CGA acts as anticancer molecule affecting the cell cycle and provoking cell growth inhibition mainly by apoptosis induction. We also provide evidence that CGA strongly activates extracellular-signal-regulated kinase1/2 (ERK1/2). Strikingly, ERK1/2 inhibitor PD98059 sensitizes the cells to CGA. Altogether, our data enforce the evidence of the anticancer activity mediated by CGA and provide the rationale for the development of innovative therapeutic strategies in OS cure.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Chlorogenic Acid/pharmacology , Osteosarcoma/drug therapy , Adolescent , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Child , Drug Resistance, Neoplasm/drug effects , Female , Flavonoids/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Male , Mice , Middle Aged , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , NIH 3T3 Cells/drug effects , Osteosarcoma/genetics , Osteosarcoma/pathology
2.
Biochem Pharmacol ; 152: 104-113, 2018 06.
Article in English | MEDLINE | ID: mdl-29574069

ABSTRACT

Triple negative breast cancer (TNBC) is an invasive, metastatic, highly aggressive tumor. Cytotoxic chemotherapy represents the current treatment for TNBC. However, relapse and chemo-resistance are very frequent. Therefore, new therapeutic approaches that are able to increase the sensitivity to cytotoxic drugs are needed. Forskolin, a natural cAMP elevating agent, has been used for several centuries in medicine and its safeness has also been demonstrated in modern studies. Recently, forskolin is emerging as a possible novel molecule for cancer therapy. Here, we investigate the effects of forskolin on the sensitivity of MDA-MB-231 and MDA-MB-468 TNBC cells to doxorubicin through MTT assay, flow cytometry-based assays (cell-cycle progression and cell death), cell number counting and immunoblotting experiments. We demonstrate that forskolin strongly enhances doxorubicin-induced antiproliferative effects by cell death induction. Similar effects are observed with IBMX and isoproterenol cAMP elevating agents and 8-Br-cAMP analog, but not by using 8-pCPT-2'-O-Me-cAMP Epac activator. It is important to note that the forskolin-induced potentiation of sensitivity to doxorubicin is accompanied by a strong inhibition of ERK1/2 phosphorylation, is mimicked by ERK inhibitor PD98059 and is prevented by pre-treatment with Protein Kinase A (PKA) and adenylate cyclase inhibitors. Altogether, our data indicate that forskolin sensitizes TNBC cells to doxorubicin via a mechanism depending on the cAMP/PKA-mediated ERK inhibition. Our findings sustain the evidence of anticancer activity mediated by forskolin and encourage the design of future in-vivo/clinical studies in order to explore forskolin as a doxorubicin sensitizer for possible use in TNBC patients.


Subject(s)
Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Triple Negative Breast Neoplasms/drug therapy , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/genetics , Doxorubicin , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Flavonoids/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Protein Kinase Inhibitors/pharmacology
3.
Mol Biosyst ; 13(6): 1246, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28524204

ABSTRACT

Correction for 'Metabolomic profiling and biochemical evaluation of the follicular fluid of endometriosis patients' by Marianna Santonastaso et al., Mol. BioSyst., 2017, DOI: 10.1039/c7mb00181a.

4.
J Cell Physiol ; 232(5): 922-927, 2017 05.
Article in English | MEDLINE | ID: mdl-27739063

ABSTRACT

Cancer is a major public health problem and the second leading cause of mortality around the world. Although continuous advances in the science of oncology and cancer research are now leading to improved outcomes for many cancer patients, novel cancer treatment options are strongly demanded. Naturally occurring compounds from a variety of vegetables, fruits, and medicinal plants have been shown to exhibit various anticancer properties in a number of in vitro and in vivo studies and represent an attractive research area for the development of new therapeutic strategies to fight cancer. Forskolin is a diterpene produced by the roots of the Indian plant Coleus forskohlii. The natural compound forskolin has been used for centuries in traditional medicine and its safety has also been documented in conventional modern medicine. Forskolin directly activates the adenylate cyclase enzyme, that generates cAMP from ATP, thus, raising intracellular cAMP levels. Notably, cAMP signaling, through the PKA-dependent and/or -independent pathways, is very relevant to cancer and its targeting has shown a number of antitumor effects, including the induction of mesenchymal-to-epithelial transition, inhibition of cell growth and migration and enhancement of sensitivity to conventional antitumor drugs in cancer cells. Here, we describe some features of cAMP signaling that are relevant to cancer biology and address the state of the art concerning the natural cAMP elevating compound forskolin and its perspectives as an effective anticancer agent. J. Cell. Physiol. 232: 922-927, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Colforsin/therapeutic use , Cyclic AMP/metabolism , Neoplasms/drug therapy , Animals , Colforsin/chemistry , Colforsin/pharmacology , Humans , Models, Biological , Neoplasms/pathology , Signal Transduction/drug effects
5.
Molecules ; 20(9): 15910-28, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26340617

ABSTRACT

Due to its expression profile, triple-negative breast cancer (TNBC) is refractory to the most effective targeted therapies available for breast cancer treatment. Thus, cytotoxic chemotherapy represents the mainstay of treatment for early and metastatic TNBC. Therefore, it would be greatly beneficial to develop therapeutic approaches that cause TNBC cells to increase their sensitivity to cytotoxic drugs. Inorganic phosphate (Pi) is emerging as an important signaling molecule in many cell types. Interestingly, it has been shown that Pi greatly enhances the sensitivity of human osteosarcoma cell line (U2OS) to doxorubicin. We investigated the effects of Pi on the sensitivity of TNBC cells to doxorubicin and the underlying molecular mechanisms, carrying out flow cytometry-based assays of cell-cycle progression and cell death, MTT assays, direct cell number counting and immunoblotting experiments. We report that Pi inhibits the proliferation of triple-negative MDA-MB-231 breast cancer cells mainly by slowing down cell cycle progression. Interestingly, we found that Pi strongly increases doxorubicin-induced cytotoxicity in MDA-MB-231 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 dynamically accompanied by profound changes in Erk1/2 and Stat3 protein and phosphorylation levels. Altogether, our data enforce the evidence of Pi acting as a signaling molecule in MDA-MB-231 cells, capable of inhibiting Erk and Stat3 pathways and inducing sensitization to doxorubicin of TNBC cells, and suggest that targeting Pi levels at local sites might represent the rationale for developing effective and inexpensive strategies for improving triple-negative breast cancer therapy.


Subject(s)
Doxorubicin/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphates/pharmacology , STAT3 Transcription Factor/metabolism , Breast Neoplasms/enzymology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Signal Transduction/drug effects
6.
EXCLI J ; 13: 843-55, 2014.
Article in English | MEDLINE | ID: mdl-26417307

ABSTRACT

Protein Kinase A (PKA) is a well known member of the serine-threonin protein kinase superfamily. PKA, also known as cAMP-dependent protein kinase, is a multi-unit protein kinase that mediates signal transduction of G-protein coupled receptors through its activation upon cAMP binding. The widespread expression of PKA subunit genes, and the myriad of mechanisms by which cAMP is regulated within a cell suggest that PKA signaling is one of extreme importance to cellular function. It is involved in the control of a wide variety of cellular processes from metabolism to ion channel activation, cell growth and differentiation, gene expression and apoptosis. Importantly, since it has been implicated in the initiation and progression of many tumors, PKA has been proposed as a novel biomarker for cancer detection, and as a potential molecular target for cancer therapy. Here, we highlight some features of cAMP/PKA signaling that are relevant to cancer biology and present an update on targeting PKA in cancer therapy.

7.
Trop Anim Health Prod ; 45(8): 1697-702, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23712396

ABSTRACT

Growth, weight at birth and daily weight gain (DWG) on 12 water buffalo calves, starting from 6 days of age until completion of weaning, was investigated in this study. Different feeding regimens were given to two groups of animals with regard to daily milk replacer: (1) group 1 (G1) received a double concentration in single administration; whereas (2) group 2 (G2) received the same amount of milk replacer split twice daily. Blood samples were collected from each calf on days 6, 30, 60 and 90 to evaluate acute phase proteins (haptoglobin), bactericide activity, lysozime, total protein content and biochemical parameters. No differences were observed between the two groups in terms of dry matter intake, feed efficiency and live body weight at the end of the study. Interestingly, a significantly (P < 0.05) reduced DWG was observed earlier in G1 (day 45) than in G2 (day 60). Gastrointestinal disorders were not recorded throughout the experimental period, and no significant differences were recorded between the two groups for all considered parameters. This study confirms the possibility of utilising one daily administration of milk replacer in water buffalo calf during weaning. This new approach facilitates calves management, without interfering with calves growing performances.


Subject(s)
Buffaloes/physiology , Milk/metabolism , Animals , Animals, Suckling , Body Weight , Buffaloes/blood , Buffaloes/growth & development , Buffaloes/metabolism , Eating , Feces/chemistry , Female , Haptoglobins/metabolism , Italy , Leukocytes, Mononuclear/metabolism , Muramidase/blood
8.
Oncol Rep ; 29(5): 1689-96, 2013 May.
Article in English | MEDLINE | ID: mdl-23446517

ABSTRACT

Novel therapeutic approaches are required for the treatment of osteosarcoma. Combination chemotherapy is receiving increased attention in order to identify compounds that may increase the therapeutic index of clinical anticancer drugs. In this regard, naturally occurring molecules with antitumor activity and with limited toxicity to normal tissues have been suggested as possible candidates for investigation of their synergistic efficacy in combination with antineoplastic drugs. Inorganic phosphate (Pi) is an essential nutrient for living organisms. Relevantly, Pi has emerged as an important signaling molecule capable of modulating multiple cellular functions by altering signal transduction pathways, gene expression and protein abundance in many cell types. Previously, we showed that Pi inhibits proliferation and aggressiveness of U2OS human osteosarcoma cells and that Pi is capable of inducing sensitization of osteosarcoma cells to doxorubicin in a p53-dependent manner. In this study, we extended the role of Pi in the chemosensitivity of osteosarcoma cells to other anticancer drugs. Specifically, we report and compare the antiproliferative effects of a combination between Pi and doxorubicin, Taxol and 5-fluorouracil (5-FU) treatments. We found that Pi increases the antiproliferative response to both Taxol and doxorubicin to a similar extent. On the other hand, Pi did not potentiate the anticancer effects induced by 5-FU. These effects were paralleled by apoptosis induction and were cell cycle-dependent. The clinical significance of our data and their potential therapeutic applications for improving osteosarcoma treatment are discussed.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bone Neoplasms/drug therapy , Osteosarcoma/drug therapy , Phosphates/pharmacology , Apoptosis/drug effects , Bone Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Synergism , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Humans , Osteosarcoma/pathology , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Phosphates/administration & dosage
9.
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
10.
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
11.
Biores Open Access ; 1(6): 324-32, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23515360

ABSTRACT

Breast cancer is one of the most common malignancies and a major cause of cancer death among women worldwide. The high mortality rate associated with breast cancer is mainly due to a propensity of the tumor to metastasize, even if small or undetectable. Given the relevant role of leptin in breast cancer growth and metastasis, novel strategies to counteract biological effects of this obesity-linked cytokine are warranted. Recently, we demonstrated that in MDA-MB-231 breast cancer cells, intracellular cAMP elevation completely abrogates both ERK1/2 and STAT3 phosphorylation in response to leptin. Very surprisingly, this provided evidence that when cAMP levels are increased, leptin drives cells towards apoptosis associated with a marked decrease of Bcl2 protein levels and accompanied by down-regulation of protein kinase A (PKA). The aim of the current study was to investigate the role of cAMP in leptin-associated motility of breast cancer cells. Here we show that cAMP elevation completely prevents leptin-induced migration of MDA-MB-231 breast cancer cells. Interestingly, the inhibition by cAMP-elevating agents of leptin-mediated cell migration is accompanied by a strong decrease of ß3 integrin subunit and focal adhesion kinase (FAK) protein levels. Analysis of the underlying cAMP-dependent molecular mechanisms revealed that PKA blockers partly counteract the inhibition of leptin-induced migration and completely prevent the antiproliferative action by cAMP elevation. Moreover, a cAMP analogue that specifically activates Epac and not PKA has an inhibitory effect on leptin-induced cell migration as well. The present study confirms initial evidence for the efficacy of cAMP elevation against oncogenic effects of leptin, identifies ß3 integrin subunit and FAK as proteins strongly down-regulated by cAMP elevation, and suggests that both cAMP/PKA- and cAMP/Epac-dependent pathways are involved in inhibition of leptin-induced migration of MDA-MB-231 breast cancer cells. The potential clinical significance and therapeutic applications of our data are discussed.

12.
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
13.
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
14.
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
15.
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
16.
Interact Cardiovasc Thorac Surg ; 7(3): 419-24; discussion 424, 2008 May.
Article in English | MEDLINE | ID: mdl-18353850

ABSTRACT

B-type natriuretic peptide is synthesized in response to increased ventricular wall stress (WS) and hypertrophy. To serially evaluate amino-terminal-pro-BNP (NT-pBNP) serum levels in patients undergoing aortic valve replacement (AVR) for severe chronic aortic regurgitation (AR), blood samples were drawn preoperatively, 15 days postoperatively, at 6- and 12-month follow-up in 25 consecutive patients. Two-dimensional echocardiography was performed concomitantly, assessing left ventricular (LV) dimensional and functional parameters, including WS. Correlations between NT-pBNP, clinical and echocardiographic data were assessed by non-parametric statistics. Median preoperative NT-pro-BNP was 276 pg/ml (IQR=85-1056), being normal or mildly increased in 20 patients, overly increased in five. The most significant correlations of preoperative NT-pBNP were with diastolic (r=0.80, P<0.001) and systolic (r=0.75, P<0.001) meridional WS and inversely with time from symptom onset (r=-0.67, P=0.001). NT-pBNP increased 15 days postoperatively (568 pg/ml, P=0.006 vs. preoperative), then decreased at 6 months (144 pg/ml, P<0.001) to remain stable at 1 year (108 pg/ml, P=0.16). Long-term follow-up NT-pBNP showed direct correlation with diastolic WS (r=0.56, P=0.02). Higher preoperative levels of NT-pBNP predicted greater magnitude of total LV mass regression at follow-up (r=-0.65, P=0.002) independent of preoperative LV mass index, showing that NT-pBNP may have a potential prognostic usefulness in adjunct to echocardiography.


Subject(s)
Aortic Valve Insufficiency/blood , Aortic Valve Insufficiency/diagnostic imaging , Heart Valve Prosthesis Implantation , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Ventricular Function, Left , Adult , Aortic Valve Insufficiency/physiopathology , Aortic Valve Insufficiency/surgery , Biomarkers/blood , Chronic Disease , Female , Follow-Up Studies , Humans , Male , Middle Aged , Postoperative Period , Predictive Value of Tests , Preoperative Care , Prospective Studies , Severity of Illness Index , Time Factors , Treatment Outcome , Ultrasonography , Ventricular Remodeling
17.
J Interferon Cytokine Res ; 27(7): 589-97, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17651020

ABSTRACT

Doxorubicin (Doxo) is a widely used anticancer drug given for the treatment of leukemias, lymphomas, and solid tumors. Despite its potent antitumor effects, the cardiotoxicity of this drug limits its clinical use. The biochemical mechanisms of Doxo-induced cardiotoxicity remain unclear. Doxo has been shown to induce apoptosis in cardiomyocytes that seems to be responsible, at least in part, for Doxo cardiotoxicity. In this study, we investigated tumor necrosis factor-alpha (TNF-alpha) receptor-mediated signaling to better understand the causes of Doxo-induced cardiotoxicity. Here, we report that Doxo is a potent inducer of apoptosis in both H9c2 cardiomyocytes and U2OS osteosarcoma tumor cells, with significant differences in terms of kinetics and caspase activation between the two cell lines. Interestingly, Doxo-induced apoptosis is accompanied by relevant changes in TNF-alpha receptor levels in H9c2 cardiomyocytes but not in U2OS cells. Moreover, treatment with exogenous TNF-alpha strongly potentiates the apoptotic effect of Doxo in H9c2 cardiomyocytes but not in U2OS cells. Our findings show that the function of TNF receptors I and II is affected by Doxo to ultimately modulate apoptosis and cell survival in H9c2 cardiomyocytes, reinforcing the recent evidence of the relevant role of TNF-alpha receptor-mediated signaling in cardiotoxicity induced by anthracyclines.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Apoptosis , Doxorubicin/toxicity , Myocytes, Cardiac/drug effects , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Line , Cell Line, Tumor , Doxorubicin/pharmacology , Humans , I-kappa B Proteins/metabolism , Myocytes, Cardiac/metabolism , NF-KappaB Inhibitor alpha , Osteosarcoma/metabolism , Osteosarcoma/pathology , Rats , Receptors, Tumor Necrosis Factor/drug effects , Recombinant Proteins , Tumor Necrosis Factor-alpha/drug effects
18.
J Cell Biochem ; 98(6): 1584-96, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16552724

ABSTRACT

In order to elucidate how phosphate regulates cellular functions, we investigated the effects of inorganic phosphate (Pi) on adenylate cyclase (AC)/cyclic AMP (cAMP) axis. Here we describe that Pi treatment of human osteosarcoma U2OS cells results in a decrease of both intracellular cAMP levels and AC activity, and in a cell growth inhibition. The phosphate-triggered effects observed in U2OS cells are not a widespread phenomenon regarding all cell lines, since other cell lines screened respond differently to parallel Pi treatments. In U2OS cell line, the AC activity/cAMP downregulation is accompanied by significant variations in the levels of some membrane proteins belonging to the AC system. Remarkably, the above effects are blunted by pharmacological inhibition of sodium-dependent phosphate transport. Moreover, 8-Br-cAMP and other cAMP-elevating agents, such as IBMX and forskolin, interestingly, prevent the cell growth inhibition in response to phosphate. Our results enforce the increasing evidences of phosphate as a signaling molecule, identifying in U2OS cell line the AC/cAMP axis, as a novel-signaling pathway modulated by phosphate to ultimately affect cell growth.


Subject(s)
Adenylyl Cyclases/metabolism , Cyclic AMP/metabolism , Osteosarcoma/metabolism , Phosphates/pharmacology , Animals , Catalytic Domain , Cell Line, Tumor , Cell Proliferation , Dose-Response Relationship, Drug , Down-Regulation , GTP-Binding Proteins/metabolism , Humans , Mice , NIH 3T3 Cells , Phosphates/pharmacokinetics , Rats , Signal Transduction , Time Factors
19.
Cell Signal ; 16(11): 1229-37, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15337522

ABSTRACT

The adenylate cyclase (AC)/cyclic AMP (cAMP)/cAMP-dependent protein kinase pathway controls many biological phenomena. The ubiquitin/proteasome system, controlling the levels of many proteins, modulates important cellular processes such as cell cycle and cell growth. Here we describe a novel mechanism for AC regulation by proteasome pathway. Pharmacological inhibition of proteasome function in human osteosarcoma U2OS cells results in up-regulation of AC activity, increase of levels of alpha subunit of heterotrimeric stimulatory GTP-binding proteins (alphas) and, remarkably, also in preventing of beta-adrenergic receptor-mediated down-regulation of alphas protein levels. Accumulation of alphas protein is also accompanied by the appearance of polyubiquitinated alphas species. Our results: (1) identify alphas protein as a novel proteasome substrate in mammalian cells; (2) indicate that proteasome might play a physiological role in controlling AC/cAMP mediated pathways by modulating the levels of Galphas protein; (3) suggest a role for the proteasome also in controlling alphas-mediated signaling pathways other than those affecting AC complex.


Subject(s)
Adenylyl Cyclases/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction/physiology , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cyclic AMP/metabolism , Enzyme Inhibitors/pharmacology , Humans , Proteasome Inhibitors , Receptors, Adrenergic, beta/metabolism , Signal Transduction/drug effects , Ubiquitin/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
20.
Am J Hypertens ; 15(7 Pt 1): 638-43, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12118913

ABSTRACT

Leptin has been hypothesized to be a pathophysiologic link between obesity and cardiovascular diseases. Because the adenylate cyclase (AC) system is a main effector of beta-adrenergic receptors and leptin has been shown to modulate AC activity in other cell lines, a leptin impact on cardiac AC activity was hypothesized. Therefore, acute and chronic effects of leptin on a rat cardiac cell line (H9c2) were investigated. Leptin affected both basal (+ 13% at 30 min and -16.4% after 18 h v untreated cells) and catecholamine-stimulated AC activity (isoproterenol + leptin at 30 min or 18 h was +21% v untreated cells; norepinephrine + leptin at 30 min was +38.8% v untreated cells; and norepinephrine + leptin at 18 h was +6% v untreated cells). Thus, long-term leptin treatment was associated with a reduced AC activity and a different responsiveness to catecholamines. The AC activity on leptin treatment was accompanied by changes in levels of proteins structurally or functionally related to AC complex (AC, Gas, Gai, p21-ras). These data indicate that the AC complex is profoundly affected at more than one level by leptin treatment in the H9c2 cardiac cell line. Differences in AC activity after short- and long-term exposure to leptin and the interaction between leptin and catecholamine might provide further insight to the understanding of the development of hypertension and congestive heart failure in obese patients.


Subject(s)
Adenylyl Cyclases/metabolism , Leptin/pharmacology , Myocytes, Cardiac/enzymology , Adrenergic alpha-Agonists/pharmacology , Animals , Cardiotonic Agents/pharmacology , Cell Line , Drug Interactions , Enzyme Activation/drug effects , GTP-Binding Proteins/metabolism , Isoproterenol/pharmacology , Myocytes, Cardiac/cytology , Norepinephrine/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...