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










Database
Language
Publication year range
1.
Oncotarget ; 7(46): 74686-74700, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27732960

ABSTRACT

Selective targeting of the PML/RARα oncoprotein demonstrates a successful molecular targeted therapy in acute promyelocytic leukemia (APL) with a typical t(15:17) chromosomal translocation. The zinc-thiolate coordination is critical for structural stability of zinc finger proteins, including the PML moiety of PML/RARα. Based on the known interaction of redox-active selenium compounds with thiolate ligands of zinc, we herein have investigated the abrogatory effects of selenite alone or in combination with all-trans retinoic acid on PML/RARα and the possible effects on differentiation in these cells. At pharmacological concentrations, selenite inhibited the proliferation and survival of APL originated NB4 cells. In combination with ATRA, it potentiated the differentiation of NB4 cells without any differentiating effects of its own as a single agent. Concordant with our hypothesis, PML/RARα oncoprotein expression was completely abrogated by selenite. Increased expression of RARα, PU.1 and FOXO3A transcription factors in the combined treatment suggested the plausible basis for increased differentiation in these cells. We show that selenite at clinically achievable dose targets PML/RARα oncoprotein for degradation and potentiates differentiation of promyelocytic leukemic cells in combination with ATRA. The present investigation reveals the hitherto unknown potential of selenite in targeted abrogation of PML/RARα in APL cells with prospective therapeutic value.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Leukemia, Promyelocytic, Acute/pathology , Selenious Acid/pharmacology , Tretinoin/pharmacology , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Gene Expression Regulation, Leukemic/drug effects , Glutaredoxins/genetics , Glutaredoxins/metabolism , Humans , Leukemia, Promyelocytic, Acute/genetics , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Thioredoxins/genetics , Thioredoxins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured
2.
Nutrients ; 7(6): 4978-94, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26102212

ABSTRACT

BACKGROUND: Sodium selenite at high dose exerts antitumor effects and increases efficacy of cytostatic drugs in multiple preclinical malignancy models. We assessed the safety and efficacy of intravenous administered sodium selenite in cancer patients' refractory to cytostatic drugs in a phase I trial. Patients received first line of chemotherapy following selenite treatment to investigate altered sensitivity to these drugs and preliminary assessment of any clinical benefits. MATERIALS AND METHODS: Thirty-four patients with different therapy resistant tumors received iv sodium selenite daily for consecutive five days either for two weeks or four weeks. Each cohort consisted of at least three patients who received the same daily dose of selenite throughout the whole treatment. If 0/3 patients had dose-limiting toxicities (DLTs), the study proceeded to the next dose-level. If 2/3 had DLT, the dose was considered too high and if 1/3 had DLT, three more patients were included. Dose-escalation continued until the maximum tolerated dose (MTD) was reached. MTD was defined as the highest dose-level on which 0/3 or 1/6 patients experienced DLT. The primary endpoint was safety, dose-limiting toxic effects and the MTD of sodium selenite. The secondary endpoint was primary response evaluation. RESULTS AND CONCLUSION: MTD was defined as 10.2 mg/m(2), with a calculated median plasma half-life of 18.25 h. The maximum plasma concentration of selenium from a single dose of selenite increased in a nonlinear pattern. The most common adverse events were fatigue, nausea, and cramps in fingers and legs. DLTs were acute, of short duration and reversible. Biomarkers for organ functions indicated no major systemic toxicity. In conclusion, sodium selenite is safe and tolerable when administered up to 10.2 mg/m(2) under current protocol. Further development of the study is underway to determine if prolonged infusions might be a more effective treatment strategy.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Neoplasms/drug therapy , Sodium Selenite/pharmacokinetics , Sodium Selenite/toxicity , Administration, Intravenous , Adult , Aged , Biomarkers/blood , Dose-Response Relationship, Drug , Drug Administration Schedule , Endpoint Determination , Fatigue , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Nausea , Sodium Selenite/blood , Treatment Outcome
3.
Basic Clin Pharmacol Toxicol ; 114(5): 377-86, 2014 May.
Article in English | MEDLINE | ID: mdl-24529300

ABSTRACT

Selenium is an essential trace element with growth-modulating properties. Decades of research clearly demonstrate that selenium compounds inhibit the growth of malignant cells in diverse experimental model systems. However, the growth-modulating and cytotoxic mechanisms are diverse and far from clear. Lately, a remarkable tumour selective cytotoxicity of selenium compounds has been shown, indicating the potential of selenium in the treatment of cancer. Of particular interest are the redox-active selenium compounds exhibiting cytotoxic potential to tumour cells. These selenium compounds elicit complex patterns of pharmacodynamics and pharmacokinetics, leading to cell death pathways that differ among compounds. Modern oncology often focuses on targeted ligand-based therapeutic strategies that are specific to their molecular targets. These drugs are initially efficient, but the tumour cells often rapidly develop resistance against these drugs. In contrast, certain redox-active selenium compounds induce complex cascades of pro-death signalling at pharmacological concentrations with superior tumour specificity. The target molecules are often the ones that are important for the survival of cancer cells and often implicated in drug resistance. Therefore, the chemotherapeutic applications of selenium offer great possibilities of multi-target attacks on tumour cells. This MiniReview focuses on the tumour-specific cytotoxic effects of selenium, with special emphasis on cascades of cellular events induced by the major groups of pharmacologically active selenium compounds. Furthermore, the great pharmacological potential of selenium in the treatment of resistant cancers is discussed.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Selenium Compounds/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm , Humans , Molecular Targeted Therapy , Neoplasms/pathology , Oxidation-Reduction/drug effects , Selenium/metabolism , Selenium Compounds/pharmacology
4.
J Cell Mol Med ; 18(4): 671-84, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24400844

ABSTRACT

Selenium compounds inhibit neoplastic growth. Redox active selenium compounds are evolving as promising chemotherapeutic agents through tumour selectivity and multi-target response, which are of great benefit in preventing development of drug resistance. Generation of reactive oxygen species is implicated in selenium-mediated cytotoxic effects on cancer cells. Recent findings indicate that activation of diverse intracellular signalling leading to cell death depends on the chemical form of selenium applied and/or cell line investigated. In the present study, we aimed at deciphering different modes of cell death in a single cell line (HeLa) upon treatment with three redox active selenium compounds (selenite, selenodiglutathione and seleno-DL-cystine). Both selenite and selenodiglutathione exhibited equipotent toxicity (IC50 5 µM) in these cells with striking differences in toxicity mechanisms. Morphological and molecular alterations provided evidence of necroptosis-like cell death in selenite treatment, whereas selenodiglutathione induced apoptosis-like cell death. We demonstrate that selenodiglutathione efficiently glutathionylated free protein thiols, which might explain the early differences in cytotoxic effects induced by selenite and selenodiglutathione. In contrast, seleno-DL-cystine treatment at an IC50 concentration of 100 µM induced morphologically two distinct different types of cell death, one with apoptosis-like phenotype, while the other was reminiscent of paraptosis-like cell death, characterized by induction of unfolded protein response, ER-stress and occurrence of large cytoplasmic vacuoles. Collectively, the current results underline the diverse cytotoxic effects and variable potential of redox active selenium compounds on the survival of HeLa cells and thereby substantiate the potential of chemical species-specific usage of selenium in the treatment of cancers.


Subject(s)
Apoptosis/drug effects , Cell Survival/drug effects , Glutathione/analogs & derivatives , Organoselenium Compounds/administration & dosage , Selenium/administration & dosage , Antineoplastic Agents/administration & dosage , Glutathione/administration & dosage , HeLa Cells , Humans , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism
5.
PLoS One ; 7(11): e50727, 2012.
Article in English | MEDLINE | ID: mdl-23226364

ABSTRACT

Naturally occurring selenium compounds like selenite and selenodiglutathione are metabolized to selenide in plants and animals. This highly reactive form of selenium can undergo methylation and form monomethylated and multimethylated species. These redox active selenium metabolites are of particular biological and pharmacological interest since they are potent inducers of apoptosis in cancer cells. The mammalian thioredoxin and glutaredoxin systems efficiently reduce selenite and selenodiglutathione to selenide. The reactions are non-stoichiometric aerobically due to redox cycling of selenide with oxygen and thiols. Using LDI-MS, we identified that the addition of S-adenosylmethionine (SAM) to the reactions formed methylselenol. This metabolite was a superior substrate to both the thioredoxin and glutaredoxin systems increasing the velocities of the nonstoichiometric redox cycles three-fold. In vitro cell experiments demonstrated that the presence of SAM increased the cytotoxicity of selenite and selenodiglutathione, which could neither be explained by altered selenium uptake nor impaired extra-cellular redox environment, previously shown to be highly important to selenite uptake and cytotoxicity. Our data suggest that selenide and SAM react spontaneously forming methylselenol, a highly nucleophilic and cytotoxic agent, with important physiological and pharmacological implications for the highly interesting anticancer effects of selenium.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Glutaredoxins/metabolism , Methanol/analogs & derivatives , Organoselenium Compounds/metabolism , Selenium Compounds/metabolism , Selenium Compounds/pharmacology , Thioredoxins/metabolism , Biological Transport , Cell Line, Tumor , Cell Survival/drug effects , Cytochromes c/metabolism , Disulfides/metabolism , Glutathione/analogs & derivatives , Glutathione/metabolism , Humans , Intracellular Space/metabolism , Methanol/metabolism , Methanol/pharmacology , Methylation , Organoselenium Compounds/pharmacology , Oxidation-Reduction , Protein Binding , S-Adenosylmethionine/metabolism , S-Adenosylmethionine/pharmacology , Superoxides/metabolism , Thioredoxin-Disulfide Reductase/metabolism
6.
Biochem J ; 429(1): 85-93, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20408818

ABSTRACT

The Grx (glutaredoxin) proteins are oxidoreductases with a central function in maintaining the redox balance within the cell. In the present study, we have explored the reactions between selenium compounds and the glutaredoxin system. Selenite, GS-Se-SG (selenodiglutathione) and selenocystine were all shown to be substrates of human Grx1, implying a novel role for the glutaredoxins in selenium metabolism. During the past few years, selenium has further evolved as a potential therapeutic agent in cancer treatment, and a leading mechanism of cytotoxicity is the generation of ROS (reactive oxygen species). Both selenite and GS-Se-SG were reduced by Grx1 and Grx2 in a non-stoichiometric manner due to redox cycling with oxygen, which in turn generated ROS. The role of Grx in selenium toxicity was therefore explored. Cells were treated with the selenium compounds in combination with transient overexpression of, or small interfering RNA against, Grx1. The results demonstrated an increased viability of the cells during silencing of Grx1, indicating that Grx1 is contributing to selenium toxicity. This is in contrast with TrxR (thioredoxin reductase), which previously was shown to protect cells from selenium cytotoxicity, verifying a diverse role between Grx and TrxR in selenium-mediated cytotoxicity. Furthermore, selenium treatment led to a marked increase in protein glutathionylation and cysteinylation that potentially can influence the activity and function of several proteins within the cell.


Subject(s)
Glutaredoxins/metabolism , Selenium Compounds/metabolism , Selenium/metabolism , Selenium/toxicity , Signal Transduction/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cytotoxins/metabolism , Cytotoxins/toxicity , Humans , Substrate Specificity/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...