Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Cell Mol Life Sci ; 71(3): 499-516, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23771630

ABSTRACT

Low delivery of many anticancer drugs across the blood-brain barrier (BBB) is a limitation to the success of chemotherapy in glioblastoma. This is because of the high levels of ATP-binding cassette transporters like P-glycoprotein (Pgp/ABCB1), which effluxes drugs back to the bloodstream. Temozolomide is one of the few agents able to cross the BBB; its effects on BBB cells permeability and Pgp activity are not known. We found that temozolomide, at therapeutic concentration, increased the transport of Pgp substrates across human brain microvascular endothelial cells and decreased the expression of Pgp. By methylating the promoter of Wnt3 gene, temozolomide lowers the endogenous synthesis of Wnt3 in BBB cells, disrupts the Wnt3/glycogen synthase kinase 3/ß-catenin signaling, and reduces the binding of ß-catenin on the promoter of mdr1 gene, which encodes for Pgp. In co-culture models of BBB cells and human glioblastoma cells, pre-treatment with temozolomide increases the delivery, cytotoxicity, and antiproliferative effects of doxorubicin, vinblastine, and topotecan, three substrates of Pgp that are usually poorly delivered across BBB. Our work suggests that temozolomide increases the BBB permeability of drugs that are normally effluxed by Pgp back to the bloodstream. These findings may pave the way to new combinatorial chemotherapy schemes in glioblastoma.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/pharmacology , Blood-Brain Barrier/metabolism , Capillary Permeability/drug effects , Dacarbazine/analogs & derivatives , Gene Expression Regulation/drug effects , Signal Transduction/drug effects , Wnt3 Protein/metabolism , Cell Line, Tumor , DNA Methylation/drug effects , DNA Methylation/genetics , Dacarbazine/pharmacology , Gene Expression Regulation/physiology , Humans , Promoter Regions, Genetic/genetics , Signal Transduction/physiology , Temozolomide , beta Catenin/metabolism
2.
Neuro Oncol ; 15(11): 1502-17, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23897632

ABSTRACT

BACKGROUND: Glioblastoma multiforme stem cells display a highly chemoresistant phenotype, whose molecular basis is poorly known. We aim to clarify this issue and to investigate the effects of temozolomide on chemoresistant stem cells. METHODS: A panel of human glioblastoma cultures, grown as stem cells (neurospheres) and adherent cells, was used. RESULTS: Neurospheres had a multidrug resistant phenotype compared with adherent cells. Such chemoresistance was overcome by apparently noncytotoxic doses of temozolomide, which chemosensitized glioblastoma cells to doxorubicin, vinblastine, and etoposide. This effect was selective for P-glycoprotein (Pgp) substrates and for stem cells, leading to an investigation of whether there was a correlation between the expression of Pgp and the activity of typical stemness pathways. We found that Wnt3a and ABCB1, which encodes for Pgp, were both highly expressed in glioblastoma stem cells and reduced by temozolomide. Temozolomide-treated cells had increased methylation of the cytosine-phosphate-guanine islands in the Wnt3a gene promoter, decreased expression of Wnt3a, disrupted glycogen synthase-3 kinase/ß-catenin axis, reduced transcriptional activation of ABCB1, and a lower amount and activity of Pgp. Wnt3a overexpression was sufficient to transform adherent cells into neurospheres and to simultaneously increase proliferation and ABCB1 expression. On the contrary, glioblastoma stem cells silenced for Wnt3a lost the ability to form neurospheres and reduced at the same time the proliferation rate and ABCB1 levels. CONCLUSIONS: Our work suggests that Wnt3a is an autocrine mediator of stemness, proliferation, and chemoresistance in human glioblastoma and that temozolomide may chemosensitize the stem cell population by downregulating Wnt3a signaling.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/metabolism , Dacarbazine/analogs & derivatives , Glioblastoma/metabolism , Wnt Signaling Pathway/drug effects , Wnt3A Protein/genetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Down-Regulation , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Temozolomide , Tumor Cells, Cultured , Wnt3A Protein/metabolism , beta Catenin/metabolism
3.
PLoS One ; 8(4): e60975, 2013.
Article in English | MEDLINE | ID: mdl-23593363

ABSTRACT

Durable tumor cell eradication by chemotherapy is challenged by the development of multidrug-resistance (MDR) and the failure to induce immunogenic cell death. The aim of this work was to investigate whether MDR and immunogenic cell death share a common biochemical pathway eventually amenable to therapeutic intervention. We found that mevalonate pathway activity, Ras and RhoA protein isoprenylation, Ras- and RhoA-downstream signalling pathway activities, Hypoxia Inducible Factor-1alpha activation were significantly higher in MDR+ compared with MDR- human cancer cells, leading to increased P-glycoprotein expression, and protection from doxorubicin-induced cytotoxicity and immunogenic cell death. Zoledronic acid, a potent aminobisphosphonate targeting the mevalonate pathway, interrupted Ras- and RhoA-dependent downstream signalling pathways, abrogated the Hypoxia Inducible Factor-1alpha-driven P-glycoprotein expression, and restored doxorubicin-induced cytotoxicity and immunogenic cell death in MDR+ cells. Immunogenic cell death recovery was documented by the ability of dendritic cells to phagocytise MDR+ cells treated with zoledronic acid plus doxorubicin, and to recruit anti-tumor cytotoxic CD8+ T lymphocytes. These data indicate that MDR+ cells have an hyper-active mevalonate pathway which is targetable with zoledronic acid to antagonize their ability to withstand chemotherapy-induced cytotoxicity and escape immunogenic cell death.


Subject(s)
Diphosphonates/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Imidazoles/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Death/drug effects , Cell Death/immunology , Cell Line, Tumor , Cholesterol/biosynthesis , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mevalonic Acid/metabolism , Models, Biological , Phagocytosis/drug effects , Prenylation/drug effects , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Terpenes/metabolism , Zoledronic Acid , ras Proteins/metabolism , rho-Associated Kinases/metabolism
4.
Biochem J ; 447(2): 301-11, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22845468

ABSTRACT

Cardioactive glycosides exert positive inotropic effects on cardiomyocytes through the inhibition of Na(+)/K(+)-ATPase. We showed previously that in human hepatoma cells, digoxin and ouabain increase the rate of the mevalonate cascade and therefore have Na(+)/K(+)-ATPase-independent effects. In the present study we found that they increase the expression and activity of 3-hydroxy-3 methylglutaryl-CoA reductase and the synthesis of cholesterol in cardiomyocytes, their main target cells. Surprisingly this did not promote intracellular cholesterol accumulation. The glycosides activated the liver X receptor transcription factor and increased the expression of ABCA1 (ATP-binding cassette protein A1) transporter, which mediates the efflux of cholesterol and its delivery to apolipoprotein A-I. By increasing the synthesis of ubiquinone, another derivative of the mevalonate cascade, digoxin and ouabain simultaneously enhanced the rate of electron transport in the mitochondrial respiratory chain and the synthesis of ATP. Mice treated with digoxin showed lower cholesterol and higher ubiquinone content in their hearts, and a small increase in their serum HDL (high-density lipoprotein) cholesterol. The results of the present study suggest that cardioactive glycosides may have a role in the reverse transport of cholesterol and in the energy metabolism of cardiomyocytes.


Subject(s)
Adenosine Triphosphate/biosynthesis , Cholesterol/biosynthesis , Digoxin/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Orphan Nuclear Receptors/physiology , Ouabain/pharmacology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoprotein A-I/metabolism , Cell Line , Electron Transport/drug effects , Hydroxymethylglutaryl CoA Reductases/metabolism , Lipoproteins, HDL/metabolism , Liver X Receptors , Male , Mevalonic Acid/metabolism , Mice , Rats , Ubiquinone/biosynthesis
5.
Blood ; 120(16): 3271-9, 2012 Oct 18.
Article in English | MEDLINE | ID: mdl-22932792

ABSTRACT

The role of Vγ9Vδ2 T cells in chronic lymphocytic leukemia (CLL) is unexplored, although these cells have a natural inclination to react against B-cell malignancies. Proliferation induced by zoledronic acid was used as a surrogate of γδ TCR-dependent stimulation to functionally interrogate Vγ9Vδ2 T cells in 106 untreated CLL patients. This assay permitted the identification of responder and low-responder (LR) patients. The LR status was associated with greater baseline counts of Vγ9Vδ2 T cells and to the expansion of the effector memory and terminally differentiated effector memory subsets. The tumor immunoglobulin heavy chain variable region was more frequently unmutated in CLL cells of LR patients, and the mevalonate pathway, which generates Vγ9Vδ2 TCR ligands, was more active in unmutated CLL cells. In addition, greater numbers of circulating regulatory T cells were detected in LR patients. In multivariate analysis, the LR condition was an independent predictor of shorter time-to-first treatment. Accordingly, the time-to-first treatment was significantly shorter in patients with greater baseline numbers of total Vγ9Vδ2 T cells and effector memory and terminally differentiated effector memory subpopulations. These results unveil a clinically relevant in vivo relationship between the mevalonate pathway activity of CLL cells and dys-functional Vγ9Vδ2 T cells.


Subject(s)
Immunologic Memory/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphocyte Activation/immunology , Mevalonic Acid/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Bone Density Conservation Agents/pharmacology , Case-Control Studies , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Diphosphonates/pharmacology , Female , Follow-Up Studies , Gene Expression Profiling , Geranyltranstransferase/antagonists & inhibitors , Humans , Imidazoles/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Survival Rate , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/immunology , Zoledronic Acid
6.
BMC Cancer ; 12: 4, 2012 Jan 04.
Article in English | MEDLINE | ID: mdl-22217342

ABSTRACT

BACKGROUND: Invasive micropapillary carcinoma (IMPC) of the breast is a distinct and aggressive variant of luminal type B breast cancer that does not respond to neoadjuvant chemotherapy. It is characterized by small pseudopapillary clusters of cancer cells with inverted cell polarity. To investigate whether hypoxia-inducible factor-1 (HIF-1) activation may be related to the drug resistance described in this tumor, we used MCF7 cancer cells cultured as 3-D spheroids, which morphologically simulate IMPC cell clusters. METHODS: HIF-1 activation was measured by EMSA and ELISA in MCF7 3-D spheroids and MCF7 monolayers. Binding of HIF-1α to MDR-1 gene promoter and modulation of P-glycoprotein (Pgp) expression was evaluated by ChIP assay and FACS analysis, respectively. Intracellular doxorubicin retention was measured by spectrofluorimetric assay and drug cytotoxicity by annexin V-FITC measurement and caspase activity assay. RESULTS: In MCF7 3-D spheroids HIF-1 was activated and recruited to participate to the transcriptional activity of MDR-1 gene, coding for Pgp. In addition, Pgp expression on the surface of cells obtained from 3-D spheroids was increased. MCF7 3-D spheroids accumulate less doxorubicin and are less sensitive to its cytotoxic effects than MCF7 cells cultured as monolayer. Finally, HIF-1α inhibition either by incubating cells with 3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole (a widely used HIF-1α inhibitor) or by transfecting cells with specific siRNA for HIF-1α significantly decreased the expression of Pgp on the surface of cells and increased the intracellular doxorubicin accumulation in MCF7 3-D spheroids. CONCLUSIONS: MCF7 breast cancer cells cultured as 3-D spheroids are resistant to doxorubicin and this resistance is associated with an increased Pgp expression in the plasma membrane via activation of HIF-1. The same mechanism may be suggested for IMPC drug resistance.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms , Carcinoma, Papillary , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/physiology , Hypoxia-Inducible Factor 1/metabolism , Annexins/analysis , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Carcinoma, Papillary/drug therapy , Carcinoma, Papillary/metabolism , Caspases/analysis , Female , Humans , Spheroids, Cellular , Tumor Cells, Cultured
7.
Biochem Pharmacol ; 82(9): 1079-89, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21803026

ABSTRACT

The widely used anticholesterolemic drugs statins decrease the synthesis of cholesterol and the isoprenylation and activity of small G-proteins such as Ras and Rho, the effectors of which are often critical in cell proliferation. Thanks to this property, it has been hypothesized that statins may have anti-tumor activities. We investigated this issue in BALB-neuT mice, which developed Her2/neu-positive mammary cancers with 100% penetrance, and in TUBO cells, a cell line established from these tumors. Contrary to the mammary glands of BALB/c mice, the tumor tissue from BALB-neuT animals had constitutively activated Ras and ERK1/2. These were reduced by the oral administration of atorvastatin, but the statin did not prevent tumor growth in mice nor reduce the proliferation of TUBO cells, although it lowered the activity of mevalonate pathway and Ras/ERK1/2 signaling. By decreasing the mevalonate pathway-derived metabolite geranylgeranyl pyrophosphate and the RhoA/RhoA kinase signaling, atorvastatin activated NF-κB, that sustained cell proliferation. Unexpectedly Her2-positive cells were much more sensitive to the inhibition of RhoA-dependent pathways than to the suppression of Ras-dependent pathways elicited by atorvastatin. Only the simultaneous inhibition of RhoA/RhoA-kinase/NF-κB and Ras/ERK1/2 signaling allowed the statin to decrease tumor cell proliferation. Our study demonstrates that Her2-positive mammary cancers have redundant signals to sustain their proliferation and shows that statins simultaneously reduce the pro-proliferative Ras/ERK1/2 axis and activate the pro-proliferative RhoA/RhoA-kinase/NF-κB axis. The latter event dissipates the antitumor efficacy that may arise from the former one. Only the association of statins and NF-κB-targeted therapies efficiently decreased proliferation of tumor cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Heptanoic Acids/therapeutic use , Hypolipidemic Agents/therapeutic use , Mammary Neoplasms, Animal/drug therapy , Pyrroles/therapeutic use , Receptor, ErbB-2/metabolism , Animals , Atorvastatin , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/genetics , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 6/genetics , Mitogen-Activated Protein Kinase 6/metabolism , NF-kappa B/metabolism , Neoplasms, Experimental/drug therapy , Receptor, ErbB-2/genetics , Terpenes , ras Proteins/genetics , ras Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
8.
J Immunol ; 187(4): 1578-90, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21753152

ABSTRACT

Vγ9Vδ2 T cells play a major role as effector cells of innate immune responses against microbes, stressed cells, and tumor cells. They constitute <5% of PBLs but can be expanded by zoledronic acid (ZA)-treated monocytes or dendritic cells (DC). Much less is known about their ability to act as cellular adjuvants bridging innate and adaptive immunity, especially in patients with cancer. We have addressed this issue in multiple myeloma (MM), a prototypic disease with several immune dysfunctions that also affect γδ T cells and DC. ZA-treated MM DC were highly effective in activating autologous γδ T cells, even in patients refractory to stimulation with ZA-treated monocytes. ZA inhibited the mevalonate pathway of MM DC and induced the intracellular accumulation and release into the supernatant of isopentenyl pyrophosphate, a selective γδ T cell activator, in sufficient amounts to induce the proliferation of γδ T cells. Immune responses against the tumor-associated Ag survivin (SRV) by MHC-restricted, SRV-specific CD8(+) αß T cells were amplified by the concurrent activation of γδ T cells driven by autologous DC copulsed with ZA and SRV-derived peptides. Ancillary to the isopentenyl pyrophosphate-induced γδ T cell proliferation was the mevalonate-independent ZA ability to directly antagonize regulatory T cells and downregulate PD-L2 expression on the DC cell surface. In conclusion, ZA has multiple immune modulatory activities that allow MM DC to effectively handle the concurrent activation of γδ T cells and MHC-restricted CD8(+) αß antitumor effector T cells.


Subject(s)
Bone Density Conservation Agents/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Dendritic Cells/immunology , Diphosphonates/pharmacology , Imidazoles/pharmacology , Multiple Myeloma/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Cell Communication/genetics , Cell Proliferation , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Hemiterpenes/immunology , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mevalonic Acid/immunology , Monocytes/immunology , Multiple Myeloma/genetics , Organophosphorus Compounds/immunology , Programmed Cell Death 1 Ligand 2 Protein , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Zoledronic Acid
9.
Biochem J ; 439(1): 141-9, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21679161

ABSTRACT

How anti-neoplastic agents induce MDR (multidrug resistance) in cancer cells and the role of GSH (glutathione) in the activation of pumps such as the MRPs (MDR-associated proteins) are still open questions. In the present paper we illustrate that a doxorubicin-resistant human colon cancer cell line (HT29-DX), exhibiting decreased doxorubicin accumulation, increased intracellular GSH content, and increased MRP1 and MRP2 expression in comparison with doxorubicin-sensitive HT29 cells, shows increased activity of the PPP (pentose phosphate pathway) and of G6PD (glucose-6-phosphate dehydrogenase). We observed the onset of MDR in HT29 cells overexpressing G6PD which was accompanied by an increase in GSH. The G6PD inhibitors DHEA (dehydroepiandrosterone) and 6-AN (6-aminonicotinamide) reversed the increase of G6PD and GSH and inhibited MDR both in HT29-DX cells and in HT29 cells overexpressing G6PD. In our opinion, these results suggest that the activation of the PPP and an increased activity of G6PD are necessary to some MDR cells to keep the GSH content high, which is in turn necessary to extrude anticancer drugs out of the cell. We think that our data provide a new further mechanism for GSH increase and its effects on MDR acquisition.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Glucosephosphate Dehydrogenase/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Drug Resistance, Multiple/genetics , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , Glucosephosphate Dehydrogenase/genetics , Humans , Reverse Transcriptase Polymerase Chain Reaction
10.
Mol Pharm ; 8(3): 683-700, 2011 Jun 06.
Article in English | MEDLINE | ID: mdl-21491921

ABSTRACT

The most frequent drawback of doxorubicin is the onset of drug resistance, due to the active efflux through P-glycoprotein (Pgp). Recently formulations of liposome-encapsulated doxorubicin have been approved for the treatment of tumors resistant to conventional anticancer drugs, but the molecular basis of their efficacy is not known. To clarify by which mechanisms the liposome-encapsulated doxorubicin is effective in drug-resistant cancer cells, we analyzed the effects of doxorubicin and doxorubicin-containing anionic liposomal nanoparticles ("Lipodox") on the drug-sensitive human colon cancer HT29 cells and on the drug-resistant HT29-dx cells. Interestingly, we did not detect any difference in drug accumulation and toxicity between free doxorubicin and Lipodox in HT29 cells, but Lipodox was significantly more effective than doxorubicin in HT29-dx cells, which are rich in Pgp. This effect was lost in HT29-dx cells silenced for Pgp and acquired by HT29 cells overexpressing Pgp. Lipodox was less extruded by Pgp than doxorubicin and inhibited the pump activity. This inhibition was due to a double effect: the liposome shell per se altered the composition of rafts in resistant cells and decreased the lipid raft-associated amount of Pgp, and the doxorubicin-loaded liposomes directly impaired transport and ATPase activity of Pgp. The efficacy of Lipodox was not increased by verapamil and cyclosporin A and was underwent interference by colchicine. Binding assays revealed that Lipodox competed with verapamil for binding Pgp and hampered the interaction of colchicine with this transporter. Site-directed mutagenesis experiments demonstrated that glycine 185 is a critical residue for the direct inhibitory effect of Lipodox on Pgp. Our work describes novel properties of liposomal doxorubicin, investigating the molecular bases that make this formulation an inhibitor of Pgp activity and a vehicle particularly indicated against drug-resistant tumors.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Doxorubicin/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Polyethylene Glycols/pharmacology , Blotting, Western , Cell Line, Tumor , Colchicine/pharmacology , Cyclosporine/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Multiple/drug effects , Fluorescent Antibody Technique , HT29 Cells , Humans , Inhibitory Concentration 50 , Mutagenesis, Site-Directed , Verapamil/pharmacology
11.
Am J Respir Cell Mol Biol ; 45(3): 625-31, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21257924

ABSTRACT

Asbestos is a naturally occurring fibrous silicate, whose inhalation is highly related to the risk of developing malignant mesothelioma (MM), and crocidolite is one of its most oncogenic types. The mechanism by which asbestos may cause MM is unclear. We have previously observed that crocidolite in human MM (HMM) cells induces NF-κB activation and stimulates the synthesis of nitric oxide by inhibiting the RhoA signaling pathway. In primary human mesothelial cells (HMCs) and HMM cells exposed to crocidolite asbestos, coincubated or not with antioxidants, we evaluated cytotoxicity and oxidative stress induction (lipid peroxidation) and the effect of asbestos on the RhoA signaling pathway (RhoA GTP binding, Rho kinase activity, RhoA prenylation, hydroxy-3-methylglutharyl-CoA reductase activity). In this paper we show that the reactive oxygen species generated by the incubation of crocidolite with primary HMCs and three HMM cell lines mediate the inhibition of 3-hydroxy-3-methylglutharyl-CoA reductase (HMGCR). The coincubation of HMCs and HMM cells with crocidolite together with antioxidants, such as Tempol, Mn-porphyrin, and the association of superoxide dismutase and catalase, prevented the cytotoxicity and lipoperoxidation caused by crocidolite alone as well as the decrease of HMGCR activity and restored the RhoA/RhoA-dependent kinase activity and the RhoA prenylation. The same effect was observed when the oxidizing agent menadione was administrated to the cells in place of crocidolite. Such a mechanism could at least partly explain the effects exerted by crocidolite fibers in mesothelial cells.


Subject(s)
Asbestos, Crocidolite/chemistry , Epithelium/pathology , Mesothelioma/metabolism , rhoA GTP-Binding Protein/metabolism , Antioxidants/metabolism , Asbestos , Cell Line , Guanosine Triphosphate/chemistry , Humans , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation , Microscopy, Fluorescence/methods , NF-kappa B/metabolism , Oxidative Stress , Reactive Oxygen Species , Signal Transduction
12.
J Control Release ; 149(2): 196-205, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-20946921

ABSTRACT

Doxorubicin is one of the most employed anticancer drugs, but its efficacy is limited by the onset of adverse effects such as drug resistance, due to the drug efflux via P-glycoprotein (Pgp). Several factors are associated to a high Pgp activity, including the amount of cholesterol in plasma membrane, which is essential to maintain the pump function. In this work we started from the following observations: 1) the drug-resistant colon cancer HT29-dx cells had a higher content of cholesterol in plasma membrane than drug-sensitive HT29 cells and a higher activity of Pgp, which was decreased by the cholesterol-lowering agent ß-methyl-cyclodextrin; 2) HT29-dx cells showed a higher synthesis of endogenous cholesterol and a higher expression of the low-density lipoprotein receptor (LDLR); 3) the anti-cholesterolemic drug simvastatin reduced the cholesterol synthesis, increased the synthesis of LDLR and lowered the Pgp activity in resistant cells. In order to circumvent drug resistance we designed a new liposomal doxorubicin, conjugated with a recombinant LDLR-binding peptide from human apoB100: this LDL-masked doxorubicin ("apo-Lipodox") was efficiently internalized by a LDLR-driven endocytosis and induced cytotoxic effects in HT29-dx cells, reversing their drug resistance. Its efficacy was further increased by simvastatin, which up-regulates the LDLR levels and contemporarily reduces the Pgp activity, thus increasing the liposomes uptake and limiting the drug efflux. We propose that the association of liposomal doxorubicin and statins may be a future promising strategy to reverse drug-resistance in human cancer cells.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Receptors, LDL/antagonists & inhibitors , Amino Acid Sequence , Antibiotics, Antineoplastic/administration & dosage , Apolipoprotein B-100/chemistry , Apolipoprotein B-100/pharmacology , Binding Sites , Cell Culture Techniques , Cell Survival/drug effects , Cholesterol/biosynthesis , Doxorubicin/administration & dosage , HT29 Cells , Humans , Liposomes , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
13.
J Cell Mol Med ; 15(7): 1492-504, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20716130

ABSTRACT

The anticancer drug doxorubicin induces the synthesis of nitric oxide, a small molecule that enhances the drug cytotoxicity and reduces the drug efflux through the membrane pump P-glycoprotein (Pgp). Doxorubicin also induces the translocation on the plasma membrane of the protein calreticulin (CRT), which allows tumour cells to be phagocytized by dendritic cells. We have shown that doxorubicin elicits nitric oxide synthesis and CRT exposure only in drug-sensitive cells, not in drug-resistant ones, which are indeed chemo-immunoresistant. In this work, we investigate the mechanisms by which nitric oxide induces the translocation of CRT and the molecular basis of this chemo-immunoresistance. In the drug-sensitive colon cancer HT29 cells doxorubicin increased nitric oxide synthesis, CRT exposure and cells phagocytosis. Nitric oxide promoted the translocation of CRT in a guanosine monophosphate (cGMP) and actin cytoskeleton-dependent way. CRT translocation did not occur in drug-resistant HT29-dx cells, where the doxorubicin-induced nitric oxide synthesis was absent. By increasing nitric oxide with stimuli other than doxorubicin, the CRT exposure was obtained also in HT29-dx cells. Although in sensitive cells the CRT translocation was followed by the phagocytosis, in drug-resistant cells the phagocytosis did not occur despite the CRT exposure. In HT29-dx cells CRT was bound to Pgp and only by silencing the latter the CRT-operated phagocytosis was restored, suggesting that Pgp impairs the functional activity of CRT and the tumour cells phagocytosis. Our work suggests that the levels of nitric oxide and Pgp critically modulate the recognition of the tumour cells by dendritic cells, and proposes a new potential therapeutic approach against chemo-immunoresistant tumours.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Colonic Neoplasms/metabolism , HT29 Cells/metabolism , Nitric Oxide/metabolism , Phagocytosis/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Antibiotics, Antineoplastic/pharmacology , Calreticulin/metabolism , Cyclic GMP/metabolism , Cytoskeleton/metabolism , Dendritic Cells/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , HT29 Cells/drug effects , Humans , Nitric Oxide Synthase/metabolism
14.
ACS Med Chem Lett ; 2(7): 494-7, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-24900337

ABSTRACT

Products 4 and 5, obtained by conjugation of doxorubicin with nitric oxide (NO) donor nitrooxy and phenylsulfonyl furoxan moieties, respectively, accumulate in doxorubicin-resistant human colon cancer cells (HT29-dx), inducing high cytotoxicity. This behavior parallels the ability of the compounds to generate NO, detected as nitrite, in these cells. Preliminary immunoblotting studies suggest that the mechanism that underlies the cytotoxic effect could involve inhibition of cellular drug efflux due to nitration of tyrosine residues of the MRP3 protein pump.

15.
Clin Res Cardiol ; 99(9): 557-64, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20467748

ABSTRACT

This study was aimed at evaluating whether the nitric oxide (NO)/cyclic GMP (cGMP) signaling pathway is altered in platelets from patients with an acute coronary syndrome (unstable angina and acute myocardial infarction). We investigated 10 patients with unstable angina (UA), 14 with acute myocardial infarction (AMI) and 14 age and sex-matched healthy subjects. The serum markers of platelet activation (sP-selectin), inflammation (TNF-alpha and erythrocyte sedimentation rate), thrombotic state (fibrinogen) and plaque disruption were significantly higher in both UA and AMI patients compared to the healthy controls. In their platelets we assessed the cGMP levels in basal conditions and after stimulation with sodium nitroprusside (SNP), and performed Western blot analysis of homogenates to measure the expression of soluble guanylate cyclase isoforms. Basal levels of cGMP (pmol/10(10) platelets) were significantly higher in platelets from UA patients (1,097 +/- 111; p < 0.0001) and AMI (1,122 +/- 77; p < 0.0001) compared to those collected from healthy controls (497 +/- 80). The platelets of AMI patients exhibited a lack of cGMP increase after SNP stimulation in comparison with UA patients. The phosphorylation of upstream (Akt1 protein kinase alpha and endothelial NO synthase) and downstream (vasodilator-stimulated phosphoprotein, VASP) signaling proteins of the NO/cGMP pathway was investigated: serine phosphorylation in Akt1, eNOS and VASP was enhanced in platelets from UA and AMI patients when compared to controls. Furthermore, in AMI patients the inhibitors of guanylate cyclase and cGMP-dependent protein kinase did not revert the VASP phosphorylation. These data suggest that platelets from AMI patients are more resistant to SNP stimulation, not only as cGMP production, but also in terms of VASP activation. From these ex vivo results we hypothesize that the increased inflammatory state which often accompanies patients with cardiovascular diseases might promote a platelet preactivation resulting in their reduced sensitivity to NO.


Subject(s)
Acute Coronary Syndrome/metabolism , Blood Platelets/metabolism , Cyclic GMP/metabolism , Nitric Oxide/biosynthesis , Signal Transduction , Aged , Case-Control Studies , Female , Humans , Male , Middle Aged
16.
J Cell Mol Med ; 14(12): 2803-15, 2010 Dec.
Article in English | MEDLINE | ID: mdl-19818098

ABSTRACT

It is unknown whether zoledronic acid (ZA) at clinically relevant doses is active against tumours not located in bone. Mice transgenic for the activated ErbB-2 oncogene were treated with a cumulative number of doses equivalent to that recommended in human beings. A significant increase in tumour-free and overall survival was observed in mice treated with ZA. At clinically compatible concentrations, ZA modulated the mevalonate pathway and affected protein prenylation in both tumour cells and macrophages. A marked reduction in the number of tumour-associated macrophages was paralleled by a significant decrease in tumour vascularization. The local production of vascular endothelial growth factor and interleukin-10 was drastically down-regulated in favour of interferon-γ production. Peritoneal macrophages and tumour-associated macrophages of ZA-treated mice recovered a full M1 antitumoral phenotype, as shown by nuclear translocation of nuclear factor kB, inducible nitric oxide synthase expression and nitric oxide production. These data indicate that clinically achievable doses of ZA inhibit spontaneous mammary cancerogenesis by targeting the local microenvironment, as shown by a decreased tumour vascularization, a reduced number of tumour-associated macrophages and their reverted polarization from M2 to M1 phenotype.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/drug effects , Diphosphonates/pharmacology , Imidazoles/pharmacology , Macrophages/drug effects , Mammary Neoplasms, Animal/drug therapy , Mevalonic Acid/metabolism , Animals , Antineoplastic Agents/administration & dosage , Diphosphonates/administration & dosage , Female , Genes, erbB-2 , Imidazoles/administration & dosage , Interferon-gamma/metabolism , Interleukin-10/metabolism , Macrophages/immunology , Macrophages/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Metabolic Networks and Pathways , Mice , Mice, Inbred BALB C , Mice, Transgenic , NF-kappa B/metabolism , Neovascularization, Pathologic , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Nitric Oxide Synthase/genetics , Protein Prenylation , Vascular Endothelial Growth Factor A/metabolism , Zoledronic Acid
17.
Fertil Steril ; 93(7): 2437-40, 2010 May 01.
Article in English | MEDLINE | ID: mdl-19819434

ABSTRACT

In swim-up-selected spermatozoa of 38 normozoospermic patients, capacitated spermatozoa exhibited enhanced pentose phosphate pathway (PPP) activity and increased expression of glucose-6-phosphate dehydrogenase (G6PD). The G6PD inhibitor DHEA and the inhibitors of NADPH oxidase apocynin and diphenylene iodonium (DPI) prevented both superoxide generation and capacitation in human spermatozoa, but whereas DPI and DHEA inhibited PPP, apocynin did not influence it, suggesting that PPP activation during capacitation is not a response to increased oxidative stress but exerts a role by supplying reducing equivalents to oxygen.


Subject(s)
Pentose Phosphate Pathway/physiology , Sperm Capacitation/physiology , Spermatozoa/metabolism , Acetophenones/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Glucose-6-Phosphate/metabolism , Glucosephosphate Dehydrogenase/genetics , Glucosephosphate Dehydrogenase/metabolism , Humans , Male , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Onium Compounds/pharmacology , Pentose Phosphate Pathway/drug effects , Pentose Phosphate Pathway/genetics , Semen Analysis , Sperm Capacitation/drug effects , Sperm Capacitation/genetics , Spermatozoa/drug effects , Spermatozoa/physiology
18.
Mol Cancer ; 8: 108, 2009 Nov 19.
Article in English | MEDLINE | ID: mdl-19925669

ABSTRACT

BACKGROUND: Doxorubicin is one of the few chemotherapeutic drugs able to exert both cytotoxic and pro-immunogenic effects against cancer cells. Following the drug administration, the intracellular protein calreticulin is translocated with an unknown mechanism onto the plasma membrane, where it triggers the phagocytosis of tumour cells by dendritic cells. Moreover doxorubicin up-regulates the inducible nitric oxide (NO) synthase (iNOS) gene in cancer cells, leading to huge amounts of NO, which in turn acts as a mediator of the drug toxicity and as a chemosensitizer agent in colon cancer. Indeed by nitrating tyrosine on the multidrug resistance related protein 3, NO decreases the doxorubicin efflux from tumour cells and enhances the drug toxicity. It is not clear if NO, beside playing a role in chemosensitivity, may also play a role in doxorubicin pro-immunogenic effects. To clarify this issue, we compared the doxorubicin-sensitive human colon cancer HT29 cells with the drug-resistant HT29-dx cells and the HT29 cells silenced for iNOS (HT29 iNOS-). RESULTS: In both HT29-dx and HT29 iNOS- cells, doxorubicin did not induce NO synthesis, had a lower intracellular accumulation and a lower toxicity. Moreover the drug failed to promote the translocation of calreticulin and the phagocytosis of HT29-dx and HT29 iNOS-cells, which resulted both chemoresistant and immunoresistant. However, if NO levels were exogenously increased by sodium nitroprusside, the chemosensitivity to doxorubicin was restored in HT29 iNOS-cells. In parallel the NO donor per se was sufficient to induce the exposure of calreticulin and to increase the phagocytosis of HT29 iNOS- cells by DCs and their functional maturation, thus mimicking the pro-immunogenic effects exerted by doxorubicin in the parental drug-sensitive HT29 cells. CONCLUSION: Our data suggest that chemo- and immuno-resistance to anthracyclines are associated in colon cancer cells and rely on a common mechanism, that is the inability of doxorubicin to induce iNOS. Therefore NO donors might represent a promising strategy to restore both chemosensitivity and immunosensitivity to doxorubicin in resistant cells.


Subject(s)
Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Doxorubicin/pharmacology , Nitric Oxide Synthase Type II/metabolism , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Calreticulin/metabolism , Cell Death/drug effects , Cell Line, Tumor , Colonic Neoplasms/enzymology , Cyclic N-Oxides/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Doxorubicin/toxicity , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Gene Silencing/drug effects , HT29 Cells , Humans , Imidazoles/pharmacology , Intracellular Space/drug effects , Intracellular Space/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Nitric Oxide/biosynthesis , Nitrites/metabolism , Nitroprusside/pharmacology , Phagocytosis/drug effects , Protein Transport/drug effects
19.
Toxicol Appl Pharmacol ; 240(3): 385-92, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19647009

ABSTRACT

Digoxin and ouabain are cardioactive glycosides, which inhibit the Na+/K+-ATPase pump and in this way they increase the intracellular concentration of cytosolic calcium ([Ca2+](i)). They are also strong inducers of the P-glycoprotein (Pgp), a transmembrane transporter which extrudes several drugs, including anticancer agents like doxorubicin. An increased amount of Pgp limits the absorption of drugs through epithelial cells, thus inducing resistance to chemotherapy. The mechanism by which cardioactive glycosides increase Pgp is not known and in this work we investigated whether digoxin and ouabain elicited the expression of Pgp with a calcium-driven mechanism. In human colon cancer HT29 cells both glycosides increased the [Ca2+](i) and this event was dependent on the calcium influx via the Na+/Ca2+ exchanger. The increased [Ca2+](i) enhanced the activity of the calmodulin kinase II enzyme, which in turn activated the transcription factor hypoxia-inducible factor-1alpha. This one was responsible for the increased expression of Pgp, which actively extruded doxorubicin from the cells and significantly reduced the pro-apoptotic effect of the drug. All the effects of glycosides were prevented by inhibiting the Na+/Ca2+ exchanger or the calmodulin kinase II. This work clarified the molecular mechanisms by which digoxin and oubain induce Pgp and pointed out that the administration of cardioactive glycosides may widely affect the absorption of drugs in colon epithelia. Moreover, our results suggest that the efficacy of chemotherapeutic agent substrates of Pgp may be strongly reduced in patients taking digoxin.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Colonic Neoplasms/metabolism , Digoxin/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ouabain/pharmacology , Base Sequence , Blotting, Western , Colonic Neoplasms/enzymology , DNA Primers , Doxorubicin/pharmacology , Electrophoretic Mobility Shift Assay , Enzyme Activation , HT29 Cells , Humans , Phosphorylation , Polymerase Chain Reaction
20.
Blood ; 113(12): 2776-90, 2009 Mar 19.
Article in English | MEDLINE | ID: mdl-18845790

ABSTRACT

Anaplastic large cell lymphoma represents a subset of neoplasms caused by translocations that juxtapose the anaplastic lymphoma kinase (ALK) to dimerization partners. The constitutive activation of ALK fusion proteins leads to cellular transformation through a complex signaling network. To elucidate the ALK pathways sustaining lymphomagenesis and tumor maintenance, we analyzed the tyrosine-kinase protein profiles of ALK-positive cell lines using 2 complementary proteomic-based approaches, taking advantage of a specific ALK RNA interference (RNAi) or cell-permeable inhibitors. A well-defined set of ALK-associated tyrosine phosphopeptides, including metabolic enzymes, kinases, ribosomal and cytoskeletal proteins, was identified. Validation studies confirmed that vasodilator-stimulated phosphoprotein and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/inosine monophosphate cyclohydrolase (ATIC) associated with nucleophosmin (NPM)-ALK, and their phosphorylation required ALK activity. ATIC phosphorylation was documented in cell lines and primary tumors carrying ALK proteins and other tyrosine kinases, including TPR-Met and wild type c-Met. Functional analyses revealed that ALK-mediated ATIC phosphorylation enhanced its enzymatic activity, dampening the methotrexate-mediated transformylase activity inhibition. These findings demonstrate that proteomic approaches in well-controlled experimental settings allow the definition of informative proteomic profiles and the discovery of novel ALK downstream players that contribute to the maintenance of the neoplastic phenotype. Prediction of tumor responses to methotrexate may justify specific molecular-based chemotherapy.


Subject(s)
Hydroxymethyl and Formyl Transferases/metabolism , Lymphoma, Large-Cell, Anaplastic/enzymology , Multienzyme Complexes/metabolism , Neoplasm Proteins/metabolism , Nucleotide Deaminases/metabolism , Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Antimetabolites, Antineoplastic/pharmacology , Carbazoles/pharmacology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Humans , Hydroxymethyl and Formyl Transferases/antagonists & inhibitors , Indazoles/pharmacology , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/pathology , Methotrexate/pharmacology , Microfilament Proteins/metabolism , Molecular Sequence Data , Multienzyme Complexes/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Nucleotide Deaminases/antagonists & inhibitors , Phenylurea Compounds/pharmacology , Phosphoproteins/metabolism , Phosphorylation , Phosphotyrosine/analysis , Protein Interaction Mapping , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/antagonists & inhibitors , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...