Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Eur J Med Chem ; 122: 611-618, 2016 Oct 21.
Article in English | MEDLINE | ID: mdl-27448918

ABSTRACT

OBJECTIVE: As Selumetinib is a MEK1/2 inhibitor that has gained interest as an anti-tumor agent, the present study was designed to investigate autophagy involvement on Selumetinib-induced apoptosis in colorectal cancer (CRC) cells. METHODS: CRC cells death and cycle studies were assessed by AnnexinV-FITC and PI staining, respectively. Autophagy flux was analysed by Western Blot (LC3II and p62 protein levels) and retroviral infection of SW480 cells for siBecn1 RNA interference experiments. Confocal microscopy was used to determine mCherry-EGFP-LC3 distribution. KEY FINDINGS: The Selumetinib effects were concentration-dependent in SW480 cell line. Whereas 1 µM exerted an arrest in the cell cycle (G1 phase), higher concentrations (10 µM) induced cell death, which was accompanied by autophagy blockage in its last stages. Autophagy induction by Rapamycin (RAPA) increased cell survival, whereas pharmacology autophagy inhibition by Bafilomycin A1 (BAF), Chloroquine (CQ) or 3-Methyladenine (3-MA) increased Selumetinib-induced CRC cells death. CONCLUSIONS: Altogether, these results suggest that autophagy plays a fundamental role in CRC cells response to Selumetinib. In addition, the combination of Selumetinib with autophagy inhibitors may be a useful therapeutic strategy to enhance its activity against colorectal tumours.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Benzimidazoles/pharmacology , Colorectal Neoplasms/pathology , Antineoplastic Agents/chemistry , Benzimidazoles/chemistry , Cell Cycle/drug effects , Cell Survival/drug effects , HT29 Cells , Humans
2.
BMC Cancer ; 15: 240, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25885658

ABSTRACT

BACKGROUND: Colorectal carcinoma is a common cause of cancer. Adjuvant treatments include: 5-fluorouracil administered together with folinic acid, or more recently, oral fluoropyrimidines such as capecitabine, in combination with oxaliplatin or irinotecan. Metastatic colorectal cancer patients can benefit from other additional treatments such as cetuximab or bevacizumab. METHODS: Using cell culture techniques, we isolated clonal populations from primary cultures of ascitic effusion derived from a colon cancer patient and after several passages an established cell line, HGUE-C-1, was obtained. Genetic analysis of HGUE-C-1 cells was performed by PCR of selected exons and sequencing. Cell proliferation studies were performed by MTT assays and cell cycle analyses were performed by flow cytometry. Retinoblastoma activity was measured by luciferase assays and proteins levels and activity were analysed by Western blot or immunohistochemistry. RESULTS: We have established a new cell line from ascitic efussion of a colon cancer patient who did not respond to 5-fluorouracil or irinotecan. HGUE-C-1 cells did not show microsatellite instability and did not harbour mutations in KRAS, BRAF, PI3KCA or TP53. However, these cells showed loss of heterozygosity affecting Adenomatous Polyposis Coli and nuclear staining of ß-catenin protein. The HGUE-C-1 cell line was sensitive to erlotinib, gefitinib, NVP-BEZ235, rapamycin and trichostatin, among other drugs, but partially resistant to heat shock protein inhibitors and highly resistant to AZD-6244 and oxaliplatin, even though the patient from which this cell line was derived had not been exposed to these drugs. Molecular characterization of this cell line revealed low expression levels and activity of Retinoblastoma protein and elevated basal levels of Erk1/2 activity and p70S6K expression and activity, which may be related to chemoresistance mechanisms. CONCLUSIONS: HGUE-C-1 represents a novel and peculiar colon carcinoma model to study chemoresistance to chemotherapeutic agents and to novel anti-neoplasic drugs that interrupt signalling pathways such as the APC/ßcatenin, Ras/Raf/Mek/Erk, PI3K/mTOR/p70S6K pathways as well as histone regulation mechanisms.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Aged , Antineoplastic Agents/pharmacology , Biomarkers , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , DNA Mutational Analysis , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Gene Expression , Humans , Male , Microsatellite Instability , Mutation , Phenotype , Protein Kinase Inhibitors/pharmacology , Signal Transduction
3.
Antimicrob Agents Chemother ; 59(4): 2242-55, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25645828

ABSTRACT

Together with vector control, chemotherapy is an essential tool for the control of visceral leishmaniasis (VL), but its efficacy is jeopardized by growing resistance and treatment failure against first-line drugs. To delay the emergence of resistance, the use of drug combinations of existing antileishmanial agents has been tested systematically in clinical trials for the treatment of visceral leishmaniasis (VL). In vitro, Leishmania donovani promastigotes are able to develop experimental resistance to several combinations of different antileishmanial drugs after 10 weeks of drug pressure. Using an untargeted liquid chromatography-mass spectrometry (LC-MS) metabolomics approach, we identified metabolic changes in lines that were experimentally resistant to drug combinations and their respective single-resistant lines. This highlighted both collective metabolic changes (found in all combination therapy-resistant [CTR] lines) and specific ones (found in certain CTR lines). We demonstrated that single-resistant and CTR parasite cell lines show distinct metabolic adaptations, which all converge on the same defensive mechanisms that were experimentally validated: protection against drug-induced and external oxidative stress and changes in membrane fluidity. The membrane fluidity changes were accompanied by changes in drug uptake only in the lines that were resistant against drug combinations with antimonials, and surprisingly, drug accumulation was higher in these lines. Together, these results highlight the importance and the central role of protection against oxidative stress in the different resistant lines. Ultimately, these phenotypic changes might interfere with the mode of action of all drugs that are currently used for the treatment of VL and should be taken into account in drug development.


Subject(s)
Antiparasitic Agents/pharmacology , Drug Resistance/drug effects , Leishmania donovani/drug effects , Adaptation, Physiological , Animals , Cell Membrane/drug effects , Chromatography, High Pressure Liquid , DNA, Protozoan/genetics , Drug Combinations , Drug Resistance/genetics , Leishmania donovani/genetics , Leishmania donovani/metabolism , Mass Spectrometry , Membrane Fluidity/drug effects , Metabolomics , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
4.
Acta Histochem ; 117(1): 126-35, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25575574

ABSTRACT

This study aimed to determine whether the effects on the mouse liver caused by three periods of feeding a protein-free diet for 5 days followed by a normal complete diet for 5 days (3PFD-CD) are prevented by a constant methionine supply (3PFD+Met-CD). The expressions of carbonic anhydrase III (CAIII), fatty acid synthase (FAS), glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and glutathione S-transferase P1 (GSTP1) were assessed by proteomics and reverse transcriptase-polymerase chain reactions. The liver redox status was examined by measuring the activities of superoxide dismutase (SOD) and catalase (CAT), as well as protein carbonylation. Because oxidative stress can result in apoptosis, the activity and content of caspase-3, as well as the x-linked inhibitor of the apoptosis protein (XIAP) and mitochondrial caspase-independent apoptosis inducing factor (AIF) contents were assessed. In addition, the liver histomorphology was examined. Compared to the controls fed a normal complete diet throughout, feeding with 3PFD-CD increased the FAS content, decreased the CAIII content, decreased both the SOD and CAT activities, and increased protein carbonylation. It also activated caspase-3, decreased the XIAP content, decreased the AIF content, increased the number of GSTP1-positive foci and caspase-3-positive cells, and caused fatty livers. Conversely, the changes were lessened to varying degrees in mice fed 3PFD+Met-CD. The present results indicate that a regular Met supply lessens the biochemical changes, damage, and caspase-dependent apoptosis provoked by recurrent dietary amino acid deprivation in the mouse liver.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Methionine/pharmacology , Protein Deficiency/enzymology , Animals , Apoptosis Inducing Factor/metabolism , Female , Glutathione S-Transferase pi/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Mice , Mice, Inbred BALB C , Protein Deficiency/pathology , X-Linked Inhibitor of Apoptosis Protein/metabolism
5.
Neoplasia ; 16(10): 845-60, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25379021

ABSTRACT

Selumetinib (AZD6244, ARRY-142886) is a MEK1/2 inhibitor that has gained interest as an anti-tumour agent. We have determined the degree of sensitivity/resistance to Selumetinib in a panel of colorectal cancer cell lines using cell proliferation and soft agar assays. Sensitive cell lines underwent G1 arrest, whereas Selumetinib had no effect on the cell cycle of resistant cells. Some of the resistant cell lines showed high levels of ERK1/2 phosphorylation in the absence of serum. Selumetinib inhibited phosphorylation of ERK1/2 and RSK and had no effect on AKT phosphorylation in both sensitive and resistant cells. Furthermore, mutations in KRAS, BRAF, or PIK3CA were not clearly associated with Selumetinib resistance. Surprisingly, Selumetinib was able to inhibit phosphorylation of p70 S6 kinase (p70S6K) and its downstream target ribosomal protein S6 (RPS6) in sensitive cell lines. However, p70S6K and RPS6 phosphorylation remained unaffected or even increased in resistant cells. Moreover, in some of the resistant cell lines p70S6K and RPS6 were phosphorylated in the absence of serum. Interestingly, colorectal primary cultures derived from tumours excised to patients exhibited the same behaviour than established cell lines. Pharmacological inhibition of p70S6K using the PI3K/mTOR inhibitor NVP-BEZ235, the specific mTOR inhibitor Rapamycin and the specific p70S6K inhibitor PF-4708671 potentiated Selumetinib effects in resistant cells. In addition, biological inhibition of p70S6K using siRNA rendered responsiveness to Selumetinib in resistant cell lines. Furthermore, combination of p70S6K silencing and PF-47086714 was even more effective. We can conclude that p70S6K and its downstream target RPS6 are potential biomarkers of resistance to Selumetinib in colorectal cancer.


Subject(s)
Benzimidazoles/pharmacology , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Ribosomal Protein S6/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Class I Phosphatidylinositol 3-Kinases , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Enzyme Activation/drug effects , Humans , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Targeted Therapy , Mutation , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/drug effects , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , Quinolines/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/genetics , ras Proteins/genetics
6.
Anal Chem ; 86(19): 9807-15, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25188358

ABSTRACT

In this work, the contribution of carnosic acid (CA) and carnosol (CS), two major compounds present in rosemary, against colon cancer HT-29 cells proliferation is investigated using a comprehensive Foodomics approach. The Foodomics study reveals that CA induces transcriptional activation of genes that encode detoxifying enzymes and altered the expression of genes linked to transport and biosynthesis of terpenoids in the colon cancer cell line. Functional analysis highlighted the activation of the ROS metabolism and alteration of several genes involved in pathways describing oxidative degradation of relevant endogenous metabolites, providing new evidence about the transcriptional change induced by CA in HT-29 cells. Metabolomics analysis showed that the treatment with CA affected the intracellular levels of glutathione. Elevated levels of GSH provided additional evidence to transcriptomic results regarding chemopreventive response of cells to CA treatment. Moreover, the Foodomics approach was useful to establish the links between decreased levels of N-acetylputrescine and its degradation pathway at the gene level. The findings from this work and the predictions based on microarray data will help explore novel metabolic processes and potential signaling pathways to further elucidate the effect of CA in colon cancer cells.


Subject(s)
Abietanes/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Gene Expression Regulation, Neoplastic , Plant Extracts/pharmacology , Polyphenols/pharmacology , Rosmarinus/chemistry , Abietanes/isolation & purification , Antineoplastic Agents, Phytogenic/isolation & purification , Biological Transport/drug effects , Cell Proliferation/drug effects , Gene Expression Profiling , Glutathione/metabolism , HT29 Cells , Humans , Inactivation, Metabolic/drug effects , Metabolomics , Plant Extracts/isolation & purification , Polyphenols/isolation & purification , Putrescine/analogs & derivatives , Putrescine/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Transcription, Genetic
7.
Genes Nutr ; 8(1): 43-60, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22923011

ABSTRACT

In this work, the effect of rosemary extracts rich on polyphenols obtained using pressurized fluids was investigated on the gene expression of human SW480 and HT29 colon cancer cells. The application of transcriptomic profiling and functional enrichment analysis was done via two computational approaches, Ingenuity Pathway Analysis and Gene Set Enrichment Analysis. These two approaches were used for functional enrichment analysis as a previous step for a reliable interpretation of the data obtained from microarray analysis. Reverse transcription quantitative-PCR was used to confirm relative changes in mRNA levels of selected genes from microarrays. The selection of genes was based on their expression change, adjusted p value, and known biological function. According to genome-wide transcriptomics analysis, rosemary polyphenols altered the expression of ~4 % of the genes covered by the Affymetrix Human Gene 1.0ST chip in both colon cancer cells. However, only ~18 % of the differentially expressed genes were common to both cell lines, indicating markedly different expression profiles in response to the treatment. Differences in induction of G2/M arrest observed by rosemary polyphenols in the two colon adenocarcinoma cell lines suggest that the extract may be differentially effective against tumors with specific mutational pattern. From our results, it is also concluded that rosemary polyphenols induced a low degree of apoptosis indicating that other multiple signaling pathways may contribute to colon cancer cell death.

8.
Electrophoresis ; 33(15): 2314-27, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22887152

ABSTRACT

In this work, a global Foodomics strategy has been applied to study the antiproliferative effect of dietary polyphenols from rosemary on two human leukemia lines, one showing a drug-sensitive phenotype (K562), and another exhibiting a drug-resistant phenotype (K562/R). To this aim, whole-transcriptome microarray together with an MS-based nontargeted analytical approach (via CE-TOF MS and UPLC-TOF MS) have been employed to carry out transcriptomics and metabolomics analyses, respectively. Functional enrichment analysis was done using ingenuity pathway analysis (IPA) software as a previous step for a reliable interpretation of transcriptomic and metabolomic profiles. Rosemary polyphenols altered the expression of approximately 1% of the genes covered by the whole transcriptome microarray in both leukemia cell lines. Overall, differences in the transcriptional induction of a number of genes encoding phase II detoxifying and antioxidant genes, as well as differences in the metabolic profiles observed in the two leukemia cell lines suggest that rosemary polyphenols may exert a differential chemopreventive effect in leukemia cells with different phenotypes. IPA predictions on transcription factor analysis highlighted inhibition of Myc transcription factor function by rosemary polyphenols, which may explain the observed antiproliferative effect of rosemary extract in the leukemia cells. Metabolomics analysis suggested that rosemary polyphenols affected differently the intracellular levels of some metabolites in two leukemia cell sublines. Integration of data obtained from transcriptomics and metabolomics platforms was attempted by overlaying datasets on canonical (defined) metabolic pathways using IPA software. This strategy enabled the identification of several differentially expressed genes in the metabolic pathways modulated by rosemary polyphenols providing more evidences on the effect of these compounds.


Subject(s)
Metabolome/drug effects , Plant Extracts/pharmacology , Polyphenols/pharmacology , Rosmarinus/chemistry , Transcriptome/drug effects , Cell Cycle/drug effects , Cell Survival/drug effects , Electrophoresis, Capillary , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , K562 Cells , Metabolomics , Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
Electrophoresis ; 33(15): 2328-36, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22887153

ABSTRACT

In this study, an analytical multiplatform is presented to carry out a broad metabolomic study on the anti-proliferative effect of dietary polyphenols on human colon cancer cells. CE, RP/UPLC, and HILIC/UPLC all coupled to TOF MS were combined to achieve a global metabolomic examination of the effect of dietary polyphenols on HT29 colon cancer cells. By the use of a nontargeted metabolomic approach, metabolites showing significant different expression after the polyphenols treatment were identified in colon cancer cells. It was demonstrated that this multianalytical platform provided extensive metabolic information and coverage due to its complementary nature. Differences observed in metabolic profiles from CE-TOF MS, RP/UPLC-TOF MS, and HILIC/UPLC-TOF MS can be mainly assigned to their different separation mechanisms without discarding the influence of the different tools used for data processing. Changes in glutathione metabolism with an enhanced reduced glutathione/oxidized glutathione (GSH/GSSG) ratio were detected in polyphenols-treated cells. Moreover, significant alterations in polyamines content with important implications in cancer proliferation were observed after the treatment with polyphenols. These results from metabolomics can explain the chemopreventive effect of the tested dietary polyphenols on colon cancer and may be of importance for future prevention and/or treatment of this disease.


Subject(s)
Colonic Neoplasms/drug therapy , Metabolome/drug effects , Metabolomics/methods , Polyphenols/pharmacology , Cell Growth Processes/drug effects , Chromatography, High Pressure Liquid , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Electrophoresis, Capillary/methods , HT29 Cells , Humans , Plant Extracts/pharmacology , Rosmarinus/chemistry , Tandem Mass Spectrometry/methods
10.
BMC Mol Biol ; 13: 25, 2012 Jul 30.
Article in English | MEDLINE | ID: mdl-22846052

ABSTRACT

BACKGROUND: It has been reported that the histone deacetylase inhibitor (iHDAc) trichostatin A (TSA) induces an increase in MDR1 gene transcription (ABCB1). This result would compromise the use of iHDACs in combination with other cytotoxic agents that are substrates of P-glycoprotein (Pgp). It has also been reported the use of alternative promoters by the ABCB1 gene and the existence of a translational control of Pgp protein. Finally, the ABCB1 gene is located in a genetic locus with the nested gene RUNDC3B in the complementary DNA strand, raising the possibility that RUNDC3B expression could interfere with ABCB1 alternative promoter regulation. METHODS: A combination of RT-PCR, real time RT-PCR, Western blot and drug accumulation assays by flow cytometry has been used in this study. RESULTS: The iHDACs-induced increase in MDR1 mRNA levels is not followed by a subsequent increase in Pgp protein levels or activity in several pancreatic and colon carcinoma cell lines, suggesting a translational control of Pgp in these cell lines. In addition, the MDR1 mRNA produced in these cell lines is shorter in its 5' end that the Pgp mRNA produced in cell lines expressing Pgp protein. The different size of the Pgp mRNA is due to the use of alternative promoters. We also demonstrate that these promoters are differentially regulated by TSA. The translational blockade of Pgp mRNA in the pancreatic carcinoma cell lines could be related to alterations in the 5' end of the MDR1 mRNA in the Pgp protein expressing cell lines. In addition, we demonstrate that the ABCB1 nested gene RUNDC3B expression although upregulated by TSA is independent of the ABCB1 alternative promoter used. CONCLUSIONS: The results show that the increase in MDR1 mRNA expression after iHDACs treatment is clinically irrelevant since this mRNA does not render an active Pgp protein, at least in colon and pancreatic cancer cell lines. Furthermore, we demonstrate that TSA in fact, regulates differentially both ABCB1 promoters, downregulating the upstream promoter that is responsible for active P-glycoprotein expression. These results suggest that iHDACs such as TSA may in fact potentiate the effects of antitumour drugs that are substrates of Pgp. Finally, we also demonstrate that TSA upregulates RUNDC3B mRNA independently of the ABCB1 promoter in use.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Gene Expression Regulation/physiology , Histone Deacetylase Inhibitors/metabolism , Hydroxamic Acids/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Blotting, Western , Cell Line, Tumor , DNA Primers/genetics , Flow Cytometry , Humans , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
11.
J Chromatogr A ; 1248: 139-53, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22727325

ABSTRACT

A global methodology, called Foodomics, which allows carrying out a comprehensive evaluation of the health benefits of food ingredients is presented in this work. The new methodology is based on the combination of several analytical platforms and data processing for Transcriptomics, Proteomics and Metabolomics studies, allowing the determination of changes induced by food ingredients at molecular level. Both, the whole methodological development and its potential are presented through the investigation of a case study following a hypothesis-free strategy. Namely, the chemopreventive effect of polyphenols from rosemary was examined on the total gene, protein and metabolite expression in human HT29 colon cancer cells. Conclusions on the bioactivity of polyphenols against colon cancer cells based on the results from each single platform (Transcriptomics, Proteomics or Metabolomics) are compared with the conclusions based on the integration of the whole results from the three platforms, corroborating the interest of using a global integrative strategy as Foodomics. To our knowledge, although many papers and reviews have been published on this topic, this is the first time that Transcriptomics, Proteomics and Metabolomics platforms are put together to study the health benefits from dietary ingredients against colon cancer cells at gene, protein and metabolite level. Advantages, drawbacks and current challenges of this global analytical strategy are discussed in this work. The results from our study provide new insights on the biological mechanisms involved in the cancer risk reduction properties of dietary constituents.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Colonic Neoplasms/prevention & control , Dietary Supplements , Metabolomics/methods , Polyphenols/therapeutic use , Proteomics/methods , Rosmarinus/chemistry , Transcriptome/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Humans , Metabolome/drug effects , Phytotherapy , Polyphenols/pharmacology , Proteome/drug effects
12.
ChemMedChem ; 7(2): 301-10, 2012 Feb 06.
Article in English | MEDLINE | ID: mdl-22170592

ABSTRACT

A group of organotin(IV) complexes were prepared: [SnCy3 (DMNI)] (1), [SnCy3 (BZDO)] (2), [SnCy3 (DMFU)] (3), and [SnPh2 (BZDO)2 ] (4), for which DMNIH=2,6-dimethoxynicotinic acid, BZDOH=1,4-benzodioxane-6-carboxylic acid, and DMFUH=2,5-dimethyl-3-furoic acid. The cytotoxic activities of compounds 1-4 were tested against pancreatic carcinoma (PANC-1), erythroleukemia (K562), and two glioblastoma multiform (U87 and LN-229) human cell lines; they show very high antiproliferative activity, with IC50 values in the 150-700 nM range after incubation for 72 h. Distribution of cellular DNA upon treatment with 1-4 revealed that whereas compounds 1-3 induce apoptosis in most of the cell lines, compound 4 does not affect cell viability in any cell line tested, indicating a possible difference in cytotoxic mechanism. Studies with the daunomycin-resistant K562/R cell line expressing P-glycoprotein (Pgp) showed that compounds 1-4 are not substrates of this protein efflux pump, indicating that these compounds do not induce acquisition of multidrug resistance, which is associated with the overexpression of Pgp.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carboxylic Acids/chemistry , Coordination Complexes/chemistry , Tin/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/pharmacology , Crystallography, X-Ray , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Humans , Molecular Conformation
13.
J Cell Biochem ; 113(4): 1416-25, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22173742

ABSTRACT

The acquisition of a multidrug-resistant (MDR) phenotype by tumor cells that renders them unsusceptible to anti-neoplasic agents is one of the main causes of chemotherapy failure in human malignancies. The increased expression of P-glycoprotein (MDR1, P-gp, ABCB1) in tumor cells contributes to drug resistance by extruding chemotherapeutic agents or by regulating programmed cell death. In a study of MDR cell survival under cold stress conditions, it was found that resistant leukemic cells with P-gp over-expression, but not their sensitive counterparts, are hypersensitive to cold-induced cell death when exposed to temperatures below 4 °C. The transfection of parental cells with a P-gp-expressing plasmid makes these cells sensitive to cold stress, demonstrating an association between P-gp expression and cell death at low temperatures. Furthermore, we observed increased basal expression and activity of effector caspase-3 at physiological temperature (37 °C) in MDR cells compared with their parental cell line. Treatment with a caspase-3 inhibitor partially rescues MDR leukemic cells from cold-induced apoptosis, which suggests that the cell death mechanism may require caspase-3 activity. Taken together, these findings demonstrate that P-gp expression plays a role in MDR cell survival, and is accompanied by a collateral sensitivity to death induced by cold stress. These findings may assist in the design of specific therapeutic strategies to complement current chemotherapy treatment against cancer.


Subject(s)
Caspase 3/metabolism , Cold Temperature , Drug Resistance, Multiple , Leukemia L1210/pathology , Stress, Physiological , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Blotting, Western , Cell Death , Cell Line, Tumor , Leukemia L1210/enzymology , Leukemia L1210/metabolism , Phenotype , Phosphatidylserines/metabolism
14.
J Antimicrob Chemother ; 66(11): 2562-5, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21846675

ABSTRACT

OBJECTIVES: The present study was designed to elucidate the mechanism of tafenoquine uptake in Leishmania and its sterol dependence. METHODS: Because tafenoquine is a fluorescent compound, spectrofluorimetric analysis allowed us to monitor its uptake by Leishmania promastigotes and intracellular amastigotes, and to evaluate the effect of temperature, energy and H+ gradient on drug entry. The influence of sterols on tafenoquine uptake in Leishmania parasites was determined in experiments using sterol-depleting agents such as methyl-ß-cyclodextrin or cholesterol oxidase. RESULTS: Tafenoquine exhibited fast entry kinetics into Leishmania in an energy-independent, but pH- and temperature-dependent, non-saturable process. Furthermore, sterol depletion decreased tafenoquine uptake. CONCLUSIONS: These findings suggest that Leishmania takes up tafenoquine by a diffusion process and that decreases in membrane sterol content may induce a decrease in drug uptake.


Subject(s)
Aminoquinolines/metabolism , Antiprotozoal Agents/metabolism , Leishmania major/metabolism , Aminoquinolines/pharmacology , Antiprotozoal Agents/pharmacology , Biological Transport , Cell Membrane/metabolism , Cholesterol Oxidase/metabolism , Diffusion , Hydrogen-Ion Concentration , Leishmania major/drug effects , Leishmania major/growth & development , Sterols/metabolism , Temperature , beta-Cyclodextrins/pharmacology
15.
Electrophoresis ; 32(13): 1765-77, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21626520

ABSTRACT

In this work, four different metabolite purification approaches are investigated prior to metabolomics of human HT29 colon cancer cells. Namely, methanol deproteinization, ultrafiltration and two SPE methods using C18 and polymer-based cartridges were studied. The extracts were characterized via a metabolomic approach based on the application of CE TOF MS (CE-MS). CE-MS analysis time was less than 20 min per sample and allowed the simultaneous and reproducible analysis of more than 80 metabolites in a single run with a minimum consumption of sample and reagents. Metabolome analysis revealed in some cases important differences among the studied metabolite purification procedures. No significant differences were observed in the metabolite profile using C18 and polymer-based cartridges, or between ultrafiltration and methanol deproteinization. However, important differences were observed in the metabolomic profiles obtained from SPE and methanol deproteinization samples. These results demonstrate the crucial role of the metabolite purification strategy in metabolomics since it can bias (and in some cases mislead) the conclusions achieved by the metabolomic study.


Subject(s)
Cell Extracts/isolation & purification , Colonic Neoplasms/chemistry , Electrophoresis, Capillary/methods , Mass Spectrometry/methods , Metabolomics/methods , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HT29 Cells , Humans , Metabolome , Methanol/chemistry , Solid Phase Extraction
16.
Exp Cell Res ; 317(10): 1476-89, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21439954

ABSTRACT

Signaling through the epidermal growth factor receptor (EGFR) is relevant in glioblastoma. We have determined the effects of the EGFR inhibitor AG1478 in glioblastoma cell lines and found that U87 and LN-229 cells were very sensitive to this drug, since their proliferation diminished and underwent a marked G(1) arrest. T98 cells were a little more refractory to growth inhibition and A172 cells did not undergo a G(1) arrest. This G(1) arrest was associated with up-regulation of p27(kip1), whose protein turnover was stabilized. EGFR autophosphorylation was blocked with AG1478 to the same extent in all the cell lines. Other small-molecule EGFR tyrosine kinase inhibitors employed in the clinic, such as gefitinib, erlotinib and lapatinib, were able to abrogate proliferation of glioblastoma cell lines, which underwent a G(1) arrest. However, the EGFR monoclonal antibody, cetuximab had no effect on cell proliferation and consistently, had no effect on cell cycle either. Similarly, cetuximab did not inhibit proliferation of U87 ΔEGFR cells or primary glioblastoma cell cultures, whereas small-molecule EGFR inhibitors did. Activity of downstream signaling molecules of EGFR such as Akt and especially ERK1/2 was interrupted with EGFR tyrosine kinase inhibitors, whereas cetuximab treatment could not sustain this blockade over time. Small-molecule EGFR inhibitors were able to prevent phosphorylation of erbB3 and erbB4, whereas cetuximab only hindered EGFR phosphorylation, suggesting that EGFR tyrosine kinase inhibitors may mediate their anti-proliferative effects through other erbB family members. We can conclude that small-molecule EGFR inhibitors may be a therapeutic approach for the treatment of glioblastoma patients.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Glioblastoma/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-3/metabolism , Blotting, Western , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , ErbB Receptors/genetics , Flow Cytometry , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Immunoprecipitation , Quinazolines , RNA, Messenger/genetics , Receptor, ErbB-3/genetics , Receptor, ErbB-4 , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , Tyrphostins/pharmacology
17.
Int J Biochem Cell Biol ; 39(10): 1877-85, 2007.
Article in English | MEDLINE | ID: mdl-17588800

ABSTRACT

Previous studies have documented that while several drug-resistant cells enter apoptosis upon treatment with histone deacetylase inhibitors (iHDACs), their drug-sensitive counterparts do not. In the present study, we have investigated at the molecular level why parental drug-sensitive tumor cells do not respond to Trichostatin A and suberoylanilide hydroxamic acid, two iHDACs that promote apoptosis in drug-resistant leukaemia cells. Taking murine leukaemia L1210 cells as a model, we have determined that: (i) PKC-alpha expression is more elevated in parental L1210 than in drug-resistant L1210/R cells, (ii) activation of PKC neutralizes iHDACs-mediated apoptosis in L1210/R cells, (iii) depletion of PKC in parental L1210 cells results in a positive response to iHDACs-mediated apoptosis, and (iv) transfection of a mutant constitutively active PKC-alpha form in L1210/R cells makes the cells refractory to apoptosis induction by iHDACs. These results allow us to conclude that activation/high expression of PKC-alpha protects parental drug-sensitive L1210 cells from iHDACs-mediated apoptosis. Thus, determination of PKC-alpha levels/activity in leukaemia seems to be relevant when choosing efficient chemotherapy protocols based on the use of apoptosis-inducing anticancer drugs.


Subject(s)
Apoptosis/drug effects , Drug Resistance, Multiple/genetics , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Leukemia/genetics , Protein Kinase C-alpha/physiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , HL-60 Cells , Humans , Hydroxamic Acids/pharmacology , Leukemia/drug therapy , Mice , Mutant Proteins/genetics , Mutant Proteins/physiology , Protein Kinase C-alpha/genetics , Transfection , Tumor Cells, Cultured , Vorinostat
18.
Mol Cancer Res ; 5(6): 641-53, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17579122

ABSTRACT

The present study of inhibitors shows that the histone deacetylase-induced increase in P-glycoprotein (Pgp) mRNA (MDR1 mRNA) does not parallel either an increase in Pgp protein or an increase in Pgp activity in several colon carcinoma cell lines. Furthermore, studying the polysome profile distribution, we show a translational control of Pgp in these cell lines. In addition, we show that the MDR1 mRNA produced in these cell lines is shorter in its 5' end that the MDR1 mRNA produced in the MCF-7/Adr (human breast carcinoma) and K562/Adr (human erythroleukemia) cell lines, both of them expressing Pgp. The different size of the MDR1 mRNA is due to the use of alternative promoters. Our data suggest that the translational blockade of MDR1 mRNA in the colon carcinoma cell lines and in wild-type K562 cells could be overcome by alterations in the 5' end of the MDR1 mRNA in the resistant variant of these cell lines, as in the case of the K562/Adr cell line. This is, to our knowledge, the first report demonstrating that the presence of an additional 5' untranslated fragment in the MDR1 mRNA improves the translational efficiency of this mRNA.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/biosynthesis , Gene Expression Regulation, Neoplastic , RNA Processing, Post-Transcriptional , 5' Untranslated Regions , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Humans , K562 Cells , Models, Genetic , Nucleic Acid Conformation , Promoter Regions, Genetic , RNA, Messenger/metabolism
19.
Hepatogastroenterology ; 54(74): 377-81, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17523278

ABSTRACT

BACKGROUND/AIMS: Genetic abnormalities of the p53 gene may play a major role in the carcinogenesis of gastric stump carcinomas (GSC) and intestinal-type primary gastric carcinomas (IPGC). Also, they may modulate P-gp expression producing chemoresistance. The aim of this article is to analyze p53 genetic abnormalities and the influence of p53 gene status on P-gp expression in both types of carcinomas. METHODOLOGY: Forty-two paraffin-embedded samples of gastric carcinomas corresponding to 17 GSC and 25 IPGC were studied. P53 genetic abnormalities in exon 5-9 were screened by direct sequencing of PCR products. P53 and P-glycoprotein (P-gp) were assessed by a standard streptavidin-biotin immunoperoxidase method. Anti-p53 DO7 and anti-P-gp C494 were used as primary antibodies. RESULTS: Fourteen p53 mutations were found, 5 in GSC (29%) and 9 in IPGC (36%). Thirteen mutations were base-pair substitutions that produced a change in the amino acid sequence. Eight mutations were located at exon 7 (57%). P53 nuclear immunopositivity was observed in 12 GSC (71%) and 15 IPGC (60%). Only two carcinomas (1 IPGC and 1 GSC) harboring a p53 mutation did not show any p53 expression. All except one of the gastric carcinomas having a p53 mutation showed medium or high P-gp expression. However, there was no difference in P-gp expression between tumors with and without p53 mutation. CONCLUSIONS: The p53 genetic alterations found in GSC and IPGC could originate from a similar pathogenetic pathway. No association was demonstrated between p53 gene status and P-gp expression, although most of the carcinomas harboring a p53 mutation showed medium or high P-gp expression.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Chromosome Aberrations , Gastric Stump/pathology , Neoplasm Recurrence, Local/genetics , Stomach Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Biopsy , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA Mutational Analysis , Exons , Gene Expression Regulation, Neoplastic/physiology , Humans , Immunoenzyme Techniques , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Recurrence, Local/pathology , Stomach Neoplasms/pathology
20.
J Mol Neurosci ; 30(1-2): 121-4, 2006.
Article in English | MEDLINE | ID: mdl-17192656

ABSTRACT

Ligand-gated ion channels (LGICs) constitute an important family of complex membrane proteins acting as receptors for neurotransmitters (Barnard, 1992; Ortells and Lunt, 1995). The nicotinic acetylcholine receptor (nAChR) from Torpedo is the most extensively studied member of the LGIC family and consists of a pentameric transmembrane glycoprotein composed of four different polypeptide subunits (alpha, beta, gamma, and delta) in a 2:1:1:1 stoichiometry (Galzi and Changeux, 1995; Hucho et al., 1996) that are arranged pseudosymmetrically around a central cation-selective ion channel. Conformational transitions, from the closed (nonconducting), to agonist-induced open (ion-conducting), to desensitized (nonconducting) states, are critical for functioning of the nAChR (Karlin, 2002). The ability of the nAChR to undergo these transitions is profoundly influenced by the lipid composition of the bilayer (Barrantes, 2004). Despite existing information on lipid dependence of AChR function, no satisfactory explanation has been given on the molecular events by which specific lipids exert such effects on the activity of an integral membrane protein. To date, several hypotheses have been entertained, including (1) indirect effects of lipids through the alteration of properties of the bilayer, such as fluidity (an optimal fluidity hypothesis [Fong and McNamee, 1986]) or membrane curvature and lateral pressure (Cantor, 1997; de Kruijff, 1997), or (2) direct effects through binding of lipids to defined sites on the transmembrane portion of the protein (Jones and McNamee, 1988; Blanton and Wang, 1990; Fernández et al., 1993; Fernández-Ballester et al., 1994), which has led to the postulation of a possible role of certain lipids as peculiar allosteric ligands of the protein. In this paper we have reconstituted purified AChRs from Torpedo into complex multicomponent lipid vesicles in which the phospholipid composition has been systematically altered. Stopped-flow rapid kinetics of cation translocation and Fourier transform-infrared (FT-IR) spectroscopy studies have been used to illustrate the lipid dependence of both AChR function and AChR secondary structure, respectively.


Subject(s)
Phospholipids/pharmacology , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/physiology , Animals , Cholesterol/pharmacology , Kinetics , Membrane Lipids/pharmacology , Receptors, Nicotinic/drug effects , Spectroscopy, Fourier Transform Infrared , Torpedo
SELECTION OF CITATIONS
SEARCH DETAIL
...