Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Mol Cancer Ther ; 4(8): 1222-30, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16093438

ABSTRACT

The antitumor activity of the histone deacetylase inhibitors was tested in three well-characterized pancreatic adenocarcinoma cell lines, IMIM-PC-1, IMIM-PC-2, and RWP-1. These cell lines have been previously characterized in terms of their origin, the status of relevant molecular markers for this kind of tumor, resistance to other antineoplastic drugs, and expression of differentiation markers. In this study, we report that histone deacetylase inhibitors induce apoptosis in pancreatic cancer cell lines, independently of their intrinsic resistance to conventional antineoplastic agents. The histone deacetylase inhibitor-induced apoptosis is due to a serine protease-dependent and caspase-independent mechanism. Initially, histone deacetylase inhibitors increase Bax protein levels without affecting Bcl-2 levels. Consequently, the apoptosis-inducing factor (AIF) and Omi/HtrA2 are released from the mitochondria, with the subsequent induction of the apoptotic program. These phenomena require AIF relocalization into the nuclei to induce DNA fragmentation and a serine protease activity of Omi/HtrA2. These data, together with previous results from other cellular models bearing the multidrug resistance phenotype, suggest a possible role of the histone deacetylase inhibitors as antineoplastic agents for the treatment of human pancreatic adenocarcinoma.


Subject(s)
Adenocarcinoma/enzymology , Antineoplastic Agents/pharmacology , Apoptosis , Histone Deacetylase Inhibitors , Pancreatic Neoplasms/enzymology , Adenocarcinoma/chemistry , Apoptosis Inducing Factor , Caspases/metabolism , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Flavoproteins/analysis , Flavoproteins/metabolism , High-Temperature Requirement A Serine Peptidase 2 , Humans , Hydroxamic Acids/pharmacology , Membrane Proteins/analysis , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins , Pancreatic Neoplasms/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Serine Endopeptidases/metabolism , Vorinostat , bcl-2-Associated X Protein
13.
J Pharmacol Exp Ther ; 313(1): 112-20, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15644429

ABSTRACT

The multidrug resistance (MDR) phenotype is considered a major cause of the failure of cancer chemotherapy. The acquisition of MDR is usually mediated by the overexpression of drug efflux pumps such as glycoprotein P (P-gp) or multidrug resistance-related protein 1 (MRP1). Thus, the identification, validation, and development of compounds that mitigate the MDR phenotype by modulating the activity of these transport proteins is an important yet elusive target. Here, we have addressed this issue and screened an N-trialkylglycine-based combinatorial library composed of 5120 compounds to search for modulators of the MDR phenotype. The screening identified 20 trimers of N-alkylglycine that increased the intracellular accumulation of daunomycin (DNM) in drug-resistant L1210R tumor cells that overexpressed the P-gp. These compounds seem to act as P-gp antagonists, as evidenced by the augmentation of DNM accumulation in the L1210(P-gp) cell line, a drug-sensitive L1210 cell stably expressing the murine P-gp protein. Similarly, several of the active N-trialkylglycines also produced an increment in DNM uptake in human HL60R cells, which primarily express the MRP1 protein. Trialkylglycines notably sensitized L1210R and HL60R tumor cells to DNM with a potency that rivaled that of verapamil. These findings provide new molecular scaffolds for the development of effective chemosensitizers against the MDR phenotype that, in due turn, could be used as adjuvant drugs in cancer chemotherapy.


Subject(s)
Drug Resistance, Multiple/physiology , Oligopeptides/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Algorithms , Animals , Anthracyclines/metabolism , Calcium Channel Blockers/pharmacology , Catalysis , Cell Survival/drug effects , Combinatorial Chemistry Techniques , Daunorubicin/metabolism , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , HL-60 Cells , Humans , Mice , Models, Molecular , Multidrug Resistance-Associated Proteins/metabolism , Phenotype , Verapamil/pharmacology
14.
J Cell Biochem ; 94(1): 98-108, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15517594

ABSTRACT

It is well established that the effectiveness of anticancer drugs may result from combined cytotoxic and differentiation activities on tumor cells. Also, differentiating agents are able to alter the susceptibility of cancer cells to antineoplastic drug therapy. However, the acquisition and/or development of drug resistance that frequently appears in anticancer treatment can impair these interactions between differentiation agents and cytotoxic drugs. In the present study, we report that the acquisition of resistance to anthracyclines in two humans, promyeolocytic leukemia HL-60 and eythroleukemia K562 cell lines, results in a restricted maturation process induced by differentiating agents with respect to that exhibited by their corresponding drug-sensitive counterparts. Interestingly, differentiating agents are able to decrease the overexpression of drug-efflux pumps as it is the case of MRP1 in the resistant HL-60 cells, thus increasing the sensitivity of cells to drug treatment. In addition, susceptibility of the drug-sensitive cells to certain apoptotic stimuli is significantly reduced after differentiation. The results here reported indicate complex interactions between cytotoxic (drug therapy) and non-cytotoxic (differentiation) cancer treatments, which should be taken into account to improve therapeutic efficiency.


Subject(s)
Cell Differentiation , Drug Resistance, Neoplasm , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Promyelocytic, Acute/pathology , Blotting, Western , Cell Line, Tumor , HL-60 Cells , Humans , Reverse Transcriptase Polymerase Chain Reaction
15.
J Comb Chem ; 6(1): 135-41, 2004.
Article in English | MEDLINE | ID: mdl-14714997

ABSTRACT

The design and synthesis of a library of novel families of 3-oxopiperazinium and perhydro-3-oxo-1,4-diazepinium derivatives is reported. The library was composed of 44 3-oxopiperazinium derivatives (11 of these compounds had a spiranic skeleton) and 22 perhydro-3-oxo-1,4-diazepinium compounds. The synthetic procedure involved a 6-step sequence carried out in solution, along with the use of solid-phase linked scavengers and microwave activation for the rapid removal of the excess of amine reagents. A final cyclization step performed under mild conditions led to the charged heterocyclic moiety. Screening of this library in two biological assays identified active compounds that inhibit the activity of the vanilloid receptor TRPV1 and modulators of the multidrug resistance phenomenon. Thus, this synthetic sequence represents a facile and convenient entry to unprecedented libraries of this sort of tetraalkylammonium derivatives that may be of use for identification of novel scaffolds of diverse biological activity.


Subject(s)
Azepines/chemical synthesis , Azepines/pharmacology , Genes, MDR/drug effects , Receptors, Drug/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/metabolism , Cell Line, Tumor , Chromatography, High Pressure Liquid , Combinatorial Chemistry Techniques , Cyclization , Daunorubicin/metabolism , Drug Design , Drug Evaluation, Preclinical , Humans , Indicators and Reagents , Ion Channels/drug effects , Magnetic Resonance Spectroscopy , Mice , Oocytes , Patch-Clamp Techniques , Rats , Spectrometry, Mass, Fast Atom Bombardment , Xenopus
16.
Int J Cancer ; 104(5): 579-86, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12594812

ABSTRACT

The main goal of our study has been to analyze the efficiency of new anticancer drugs, specifically histone deacetylase inhibitors, in tumor cells bearing a multidrug resistance phenotype. We report that the histone deacetylase inhibitors, Trichostatin A and Suberoylanilide Hydroxamic Acid (SAHA), dramatically reduce cell viability and promote apoptosis in different drug-resistant cells, affecting in a much lesser extent to their parental drug-sensitive counterparts. The differential effects induced by Trichostatin A and SAHA between drug-sensitive and drug-resistant cells are reflected on the main characteristics of the resistant phenotype. Thus, reverse transcription-PCR and Western immunoblots confirm that both histone deacetylase inhibitors promote endogenous down-regulation of P-glycoprotein, which is overexpressed in the drug-resistant cells. Transfection of drug-sensitive cells with the P-glycoprotein cDNA ruled out the a priori possible association between apoptosis and down-regulation of P-glycoprotein induced by the histone deacetylase inhibitors. The results suggest a therapeutic potential of histone deacetylase inhibitors in the treatment of cancers with acquired resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Acetylation , Animals , Cell Division/drug effects , Cell Survival/drug effects , Histones/metabolism , Mice , Transfection , Tumor Cells, Cultured , Vorinostat
17.
Biochemistry ; 41(40): 12253-62, 2002 Oct 08.
Article in English | MEDLINE | ID: mdl-12356328

ABSTRACT

Purified Acetylcholine Receptor (AcChR) from Torpedo has been reconstituted at low (approximately 1:3500) and high (approximately 1:560) protein to phospholipid molar ratios into vesicles containing egg phosphatidylcholine, cholesterol, and different dimyristoyl phospholipids (dimyristoyl phosphatidylcholine, phosphatidylserine, phosphatidylglycerol and phosphatidic acid) as probes to explore the effects of the protein on phospholipid organization by differential scanning calorimetry, infrared, and fluorescence spectroscopy. All the experimental results indicate that the presence of the AcChR protein, even at the lower protein to phospholipid molar ratio, directs lateral phase separation of the monoanionic phosphoryl form of the phosphatidic acid probe, causing the formation of specific phosphatidic acid-rich lipid domains that become segregated from the bulk lipids and whose extent (phosphatidic acid sequestered into the domain, out of the total population in the vesicle) is protein-dependent. Furthermore, fluorescence energy transfer using the protein tryptophan residues as energy donors and the fluorescence probes trans-parinaric acid or diphenylhexatriene as acceptors, establishes that the AcChR is included in the domain. Other dimyristoyl phospholipid probes (phosphatidylcholine, phosphatidylserine, phosphatidylglycerol) under identical conditions could not mimic the protein-induced domain formation observed with the phosphatidic acid probe and result in ideal mixing of all lipid components in the reconstituted vesicles. Likewise, in the absence of protein, all the phospholipid probes, including phosphatidic acid, exhibit ideal mixing behavior. Since phosphatidic acid and cholesterol have been implicated in functional modulation of the reconstituted AcChR, it is suggested that such a specific modulatory role could be mediated by domain segregation of the relevant lipid classes.


Subject(s)
Phosphatidic Acids/chemistry , Receptors, Cholinergic/chemistry , Animals , Calorimetry, Differential Scanning , Fluorescence Polarization , Liposomes/chemistry , Membrane Lipids/chemistry , Protein Structure, Tertiary , Spectroscopy, Fourier Transform Infrared , Temperature , Torpedo
SELECTION OF CITATIONS
SEARCH DETAIL
...