Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
IUBMB Life ; 62(9): 688-92, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20836127

ABSTRACT

DJ-1 is a mitochondrial protein linked to Parkinson's disease. DJ-1 has been suggested to have several possible functions, although it has been mainly associated to oxidative stress defence. Changes in the two-dimensional electrophoresis pattern have been thoroughly described as a consequence of oxidative modification of the Cys106 residue. There is accumulating evidence supporting a specific role of DJ-1 in protecting dopaminergic neurons from dopamine itself. By exposing SH-SY5Y human neuroblastoma catecholaminergic cells to dopamine, we observed a specific increase in the most acidic forms in the DJ-1 two-dimensional electrophoresis pattern together with a significant decrease of the most basic spot. Unlike cells exposed to generic oxidative conditions, no additional shift was observed. The results are corroborated by a meta-analysis of the literature showing that in the absence of dopamine treatment the specific acidic form is underrepresented.


Subject(s)
Dopamine/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Neuroblastoma/metabolism , Oncogene Proteins/metabolism , Parkinson Disease , Blotting, Western , Cell Line, Tumor , Dopamine Agents/pharmacology , Electrophoresis, Gel, Two-Dimensional , Flow Cytometry , Humans , Protein Deglycase DJ-1 , Protein Isoforms/drug effects , Protein Isoforms/metabolism
2.
Biochem Pharmacol ; 80(4): 455-62, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20488164

ABSTRACT

Inappropriate activation of the IGF (insulin-like growth factor) system has been implicated in the growth and progression of a number of tumor types. Recent evidence indicates a possible role for the IGF system in modulating/mediating tumor cell response to hypoxia, a common occurrence in solid tumors, and particularly in malignant gliomas, causing tumor cells either to die, or to mount a pleiotropic adaptive response that is mainly orchestrated through activation of the hypoxia-inducible transcription factor HIF1. Experimental evidence suggests possible links between IGF- and HIF1-dependent signaling pathways, as well as a role for activated STAT3 in mediating their activities. Interestingly, igf2 is among the target genes transactivated by HIF1, thereby providing the missing link in a hypothetical autocrine self-amplifying circuit. The present study investigates the presence of the IGF-HIF1-VEGF axis in the human glioma cell line U-87 MG, and characterizes its molecular effectors. Our results show that exogenous IGF-I causes IGF1R and STAT3 activation, and increases HIF1alpha protein levels and HIF1 trascriptional activity, inducing VEGF release; a similar response, mediated by IGF-II release, is observed following HIF1alpha stabilization. The existence of an autocrine loop is confirmed by its down-regulation following inactivation of IGF1R (using the IGF1R-specific tyrosine kinase inhibitor NVP-AEW541), STAT3 (transfecting the cells with an expression vector encoding a dominant negative form of STAT3), or HIF1 (using the small molecule inhibitor YC-1). The ability of NVP-AEW541 to block this circuit could be beneficial in suppressing the growth and angiogenic potential of hypoxic glial tumors.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Glioblastoma/pathology , Insulin-Like Growth Factor I/metabolism , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
3.
Bioorg Med Chem ; 17(5): 2009-16, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19211252

ABSTRACT

Preliminary in vitro cytotoxicity studies on a panel of meso diaryl-substituted tetrapyrrole derivatives newly synthesized in our laboratory have shown that these compounds are photodynamically active on the human colon carcinoma cell line HCT116. In the present study, we investigate some mechanistic aspects of the photodynamic action of the most active compounds in the series, namely the 5-phenyl-15-(3-methoxyphenyl)porphyrin (1), the 5-phenyl-15-(3-hydroxyphenyl)porphyrin (2) and the 5,15-diphenylporphyrin (3). The results of the cytotoxicity studies indicate that the novel photosensitisers (PSs) are more potent in vitro than m-THPC (Foscan), a powerful PS already approved for clinical use in photodynamic therapy (PDT). A series of experiments were performed to elucidate a number of aspects in the mechanism of PS-induced phototoxicity, including, intracellular accumulation and subcellular localization of the PSs, induction of apoptosis, and generation of reactive oxygen species (ROS) and NO*. All the compounds tested exhibit similar singlet oxygen quantum yields; differential intracellular accumulation can contribute to the observed differences in phototoxicity. Flow cytometric studies indicate that all the tested compounds induce apoptosis; however, their cytotoxic effect does not seem to rely solely on this process. Generation of significant amounts of reactive oxygen species (ROS) and NO* were also observed; however, the contribution of this latter effect to the overall phototoxicity is unclear. Taken together, our observations suggest that the diaryl derivatives included in the present study could represent promising leads for the development of novel photosensitizing agents.


Subject(s)
Carcinoma/drug therapy , Colonic Neoplasms/drug therapy , Photosensitizing Agents/toxicity , Porphyrins/toxicity , Apoptosis , Cell Line, Tumor , Humans , Nitric Oxide/metabolism , Photochemotherapy , Photosensitizing Agents/chemistry , Porphyrins/chemistry , Reactive Oxygen Species/metabolism , Singlet Oxygen/metabolism
4.
Eur J Cancer ; 45(5): 890-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19171477

ABSTRACT

Tumour hypoxia represents a major obstacle to the success of radiotherapy and chemotherapy. The discovery that the hypoxia-inducible factor 1 (HIF-1) is a master regulator of cellular response to low oxygen led to the concept that inhibiting HIF-1 activity may sensitise hypoxic cancer cells to radiation and cytotoxic drugs. In the present study we investigate the effects of HIF-1 modulation on the response of the human colon adenocarcinoma cell line HCT116 to 5-fluorouracil (5FU). Increasing HIF-1 activity, either by exposing cells to hypoxia or by forced expression of a degradation-resistant form of HIF-1alpha, results in poor cell response to 5FU; conversely, knockdown of HIF-1alpha by RNA interference prevents hypoxia-induced resistance to 5FU. PMX290, a thioredoxin-1 inhibitor, significantly inhibits HIF-1 activity and concomitantly sensitises hypoxic cells to 5FU. These results were confirmed in HCT116 cells grown as three-dimensional spheroids, a model that more closely reproduces the hypoxic environment of solid tumours.


Subject(s)
Adenocarcinoma/pathology , Antimetabolites, Antineoplastic/pharmacology , Colonic Neoplasms/pathology , Fluorouracil/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Adenocarcinoma/metabolism , Apoptosis/drug effects , Cell Hypoxia/physiology , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Indoles/pharmacology , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , Spheroids, Cellular , Sulfones/pharmacology , Thioredoxins/antagonists & inhibitors , Thioredoxins/physiology , Tumor Cells, Cultured
5.
Oncol Rep ; 20(3): 625-30, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18695915

ABSTRACT

Essential oils from various aromatic plants have been reported to exert chemopreventive and/or antitumor effects. In addition, a number of studies have shown the ability of chemopreventive phytochemicals to increase the sensitivity of cancer cells to conventional anticancer drugs. The success of chemotherapeutic agents is often hindered by the development of drug resistance, with multidrug resistant (MDR) phenotypes reported in a number of tumors, generally involving reduced intracellular drug accumulation due to increased drug efflux by membrane transporters. In the present study, the effects of linalool (LIN), a monoterpene alcohol found in the essential oils from many aromatic plants, on the growth of two human breast adenocarcinoma cell lines, MCF7 WT and multidrug resistant MCF7 AdrR, were investigated, both as a single agent and in combination with doxorubicin (DOX). The results reported here show that LIN only moderately inhibits cell proliferation; interestingly, however, subtoxic concentrations of LIN potentiate DOX-induced cytotoxicity and pro-apoptotic effects in both cell lines. A significant synergism can be observed in MCF7 AdrR cells, which may be due, at least in part, to the ability of LIN to increase DOX accumulation and to induce a decrease in Bcl-xL levels. In summary, the results of the present study suggest that LIN may improve the therapeutic index of anthracyclines in the management of breast cancer, especially in MDR tumors.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Breast Neoplasms/drug therapy , Doxorubicin/adverse effects , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Insecticides/therapeutic use , Monoterpenes/therapeutic use , Protective Agents/therapeutic use , Acyclic Monoterpenes , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Flow Cytometry , Humans , Immunoenzyme Techniques , Inhibitor of Apoptosis Proteins , Inhibitory Concentration 50 , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Survivin , Tumor Cells, Cultured/drug effects , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
6.
Cancer Lett ; 258(2): 181-8, 2007 Dec 18.
Article in English | MEDLINE | ID: mdl-17920763

ABSTRACT

Metastatic breast cancer is an incurable disease, often characterized by poor response to standard chemotherapy, which is mainly based on anthracyclines and taxanes. Thus, increasing tumor cell sensitivity to these agents is an attractive goal towards improving the clinical management of this disease. The present study investigates the effects of signal transducer and activator of transcription 3 (Stat3) inhibition on the response of the highly metastatic MDA-MB-231 human breast adenocarcinoma cell line to doxorubicin (DOX). Stat3 is a transcription factor often constitutively activated in breast tumors and cancer cell lines, and is thought to contribute to malignant transformation and progression by transactivation of a host of target genes involved in cell proliferation and survival, angiogenesis and invasiveness. Our results indicate that (a) untreated MDA-MB-231 cells express higher baseline levels of (activated) pTyr(705)Stat3, that are further upregulated following exposure to DOX, than the non-metastatic MCF-7 cell line; (b) inhibiting the Stat3 signaling pathway, by exposure to the tyrphostin AG490 (an inhibitor of the upstream activating Janus kinases), by transfection with a dominant-negative form of Stat3 or by treatment with satraplatin (a tetravalent platinum derivative that inhibits Stat3 activation), increases breast cancer cell response to the proapoptotic effect of DOX (to different extents). In addition, the latter two approaches have been shown to interfere with expression of one or more antiapoptotic proteins. Overall, these observations suggest that suppression of Stat3 signaling may provide a potential therapeutic approach to overcoming DOX resistance in metastatic breast cancer cells.


Subject(s)
Doxorubicin/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tyrphostins/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Female , Flow Cytometry , Humans , Immunoblotting , Neoplasm Metastasis , Organoplatinum Compounds/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor/genetics , Transfection
7.
Free Radic Biol Med ; 40(8): 1409-18, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16631531

ABSTRACT

The occurrence of multidrug resistance (MDR) is the major obstacle to successful anthracycline-based cancer chemotherapy. In the present study, we assessed the effects of Tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl, TPL), a piperidine nitroxide with growth-inhibitory properties in tumor cell lines, on a number of molecular mechanisms involved in the resistance of human breast adenocarcinoma cell lines to doxorubicin (DOX). Cytotoxicity studies in MCF-7 wildtype and their MDR variant MCF-7 Adr(R) cells showed a synergistic effect between TPL and DOX when exposure to TPL preceded or was simultaneous with DOX treatment in MCF-7 Adr(R) cells. This effect of TPL seems to be due in part to its ability to increase peroxide levels and to deplete cellular glutathione pools. In addition, TPL increased DOX accumulation in MCF-7 Adr(R) cells by interfering with P-glycoprotein-mediated DOX efflux, as evidenced using a specific antibody that recognizes the active form of the protein. TPL was also found to affect the expression levels of proteins involved in response to drug treatment (e.g., p53, bcl2, bax, p21). Taken together, our results indicate that TPL is a potential new agent that may improve the clinical effect of DOX in tumors exhibiting a MDR phenotype.


Subject(s)
Breast Neoplasms/metabolism , Cyclic N-Oxides/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cell Line, Tumor , Glutathione/metabolism , Humans , Oxidative Stress , Spin Labels
8.
BMC Cancer ; 4: 92, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15601469

ABSTRACT

BACKGROUND: Colon adenocarcinomas are refractory to a number of widely used anticancer agents. Multifactorial mechanisms have been implicated in this intrinsically resistant phenotype, including deregulation of cell death pathways. In this regard, the p53 protein has a well established role in the control of tumor cell response to DNA damaging agents; however, the relationship between p53-driven genes and drug sensitivity remains controversial. The present study investigates the role of the p53/p21 system in the response of human colon carcinoma cells to treatment with the cytotoxic agent doxorubicin (DOX) and the possibility to modify the therapeutic index of DOX by modulation of p53 and/or p21 protein levels. METHODS: The relationship between p53 and p21 protein levels and the cytotoxic effect of DOX was investigated, by MTT assay and western blot analysis, in HCT116 (p53-positive) and HT29 (p53-negative) colon cancer cells. We then assessed the effects of DOX in two isogenic cell lines derived from HCT116 by abrogating the expression and/or function of p53 and p21 (HCT116-E6 and HCT116 p21-/-, respectively). Finally, we evaluated the effect of pre-treatment with the piperidine nitroxide Tempol (TPL), an agent that was reported to induce p21 expression irrespective of p53 status, on the cytotoxicity of DOX in the four cell lines. Comparisons of IC50 values and apoptotic cell percentages were performed by ANOVA and Bonferroni's test for independent samples. C.I. calculations were performed by the combination Index method. RESULTS: Our results indicate that, in the colon carcinoma cell lines tested, sensitivity to DOX is associated with p21 upregulation upon drug exposure, and DOX cytotoxicity is potentiated by pre-treatment with TPL, but only in those cell lines in which p21 can be upregulated. CONCLUSIONS: p21 induction may significantly contribute to the response of colon adenocarcinomas cells to DOX treatment; and small molecules that can exploit p53-independent pathways for p21 induction, such as TPL, may find a place in chemotherapeutic protocols for the clinical management of colorectal cancer, where p53 function is often lost, due to genetic or epigenetic defects or to post-transcriptional inactivating mechanisms.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Cell Cycle Proteins/metabolism , Colonic Neoplasms/drug therapy , Doxorubicin/therapeutic use , Neoplasm Proteins/metabolism , Tumor Suppressor Protein p53/physiology , Analysis of Variance , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Cyclic N-Oxides/therapeutic use , Cyclin-Dependent Kinase Inhibitor p21 , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Drug Synergism , HT29 Cells , Humans , Protein Synthesis Inhibitors/therapeutic use , Spin Labels , Transfection/methods
9.
Int J Oncol ; 25(6): 1817-22, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15547722

ABSTRACT

Temozolomide (TMZ) is a methylating agent with promising antitumor efficacy for the treatment of melanomas and intermediate-grade gliomas. Unfortunately, its use in the management of high-grade gliomas (glioblastomas) is limited by multifaceted resistance mechanisms. The aim of this study was to evaluate the possibility to improve the cytotoxic response of two human glioblastoma cell lines, U87MG and U373MG, to TMZ by the use of Tempol (TPL), a low molecular weight piperidine nitroxide that has been shown to inhibit in vitro and in vivo growth of murine glioma cells. To this purpose, we used two different schedules for the combined exposure to the two agents. Our data indicate that TPL synergizes with TMZ in both U87MG and U373MG cells for both schedules tested. This effect is accompanied by an increase in apoptotic cell death and by changes in the expression of genes involved in control of the apoptotic process. TPL was also observed to induce a cell-type specific decrease in GSH levels and in GSH-related enzyme activities that could contribute to its sensitizing effect.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Brain Neoplasms/pathology , Cyclic N-Oxides/pharmacology , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Glioblastoma/pathology , Cell Cycle/drug effects , Drug Interactions , Humans , Spin Labels , Temozolomide , Tumor Cells, Cultured
10.
Bioorg Med Chem ; 12(18): 4853-60, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15336264

ABSTRACT

Photodynamic therapy (PDT) is a cancer treatment involving systemic administration of a tumor-localizing photosensitizer; this, when activated by the appropriate wavelength of light, interacts with molecular oxygen to form a toxic, short-lived species known as singlet oxygen, which is thought to mediate cellular death. Photofrin, a complex mixture of porphyrin oligomers has recently received FDA approval for the photodynamic treatment of esophageal and endobronchial carcinoma, but its photodynamic and toxicity profiles are far from ideal. In the present study we evaluated a series of porphyrin-based PSs, some of which newly synthesized by our group, with the aim to identify agents with more favorable characteristics. For the most effective compounds in the porphyrin series, chlorin analogs were also synthesized; for comparison, the screening also included Photofrin. Cytotoxicity studies were performed by the MTT assay on a cultured human colon adenocarcinoma cell line (HCT116); the results indicate that the 3,4,5-trimethoxyphenyl, 3OH- and 4OH-phenyl, and the sulfonamidophenyl derivatives are significantly more potent than Photofrin. Flow cytometric studies and fluorescence microscopy indicate that in PDT-treated HCT116 cells death occurs mainly by apoptosis. In summary, novel PSs described in the present study, belonging both to the porphyrin and chlorin series, have proven more effective than Photofrin in killing colon cancer cells in vitro; extending these observation to in vivo models, particularly regarding the deeper reaching chlorin derivatives, might lead to significant advances in the development of tumor PDT.


Subject(s)
Adenocarcinoma/drug therapy , Colonic Neoplasms/drug therapy , Photosensitizing Agents/therapeutic use , Porphyrins/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dihematoporphyrin Ether/chemistry , Dihematoporphyrin Ether/pharmacology , Dihematoporphyrin Ether/therapeutic use , Dose-Response Relationship, Drug , Humans , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Porphyrins/chemistry , Porphyrins/pharmacology
11.
Int J Oncol ; 22(5): 1057-64, 2003 May.
Article in English | MEDLINE | ID: mdl-12684672

ABSTRACT

Intrinsic or acquired drug resistance poses a major challenge to the success of chemotherapy in the clinical management of human cancers. While acquired multidrug resistance (MDR), whereby cells become refractory to multiple drugs, has been extensively investigated, the mechanistic basis for intrinsic resistance remains elusive, so that this condition is largely unmanageable in the clinical setting. To address this issue, we have assessed the effects of the anticancer agent doxorubicin (DX) on a panel of human tumor cell lines originally derived from untreated patients and tried to establish a correlation between cell response and a number of parameters, including drug accumulation and/or drug efflux; differences in expression and/or subcellular distribution of proteins involved in the apoptotic process (e.g., p53, Bcl-2, Bax) and intracellular signal transducers (PKCalpha); changes in key detoxification processes. Based on our results, 'classic' multispecific drug transporters (P-glycoprotein, MDR-related proteins) only seem to play a minor role in the intrinsically resistant phenotype, whereas LRP may contribute to resistance in non-small cell lung carcinoma (NSCLC) cells. No relationship was observed between drug response and expression and/or subcellular localization of apoptosis-related proteins; however, increased PKCalpha levels are associated with poor drug response, suggesting that one or more substrates of this enzyme may be relevant to the resistant phenotype. Finally, overactive glutathione-recycling pathways may contribute to DX resistance.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Doxorubicin/pharmacokinetics , Doxorubicin/toxicity , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Apoptosis/drug effects , Biological Transport , Dose-Response Relationship, Drug , Humans , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...