Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Proteome Res ; 21(4): 940-952, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35196455

RESUMO

For more than a decade, DNA and histone methylations have been the focus of extensive work, although their relationship with methyl group metabolism was overlooked. Recently, it has emerged that epigenetic methylations are influenced by methyl donor nutrient availability, cellular levels of S-adenosyl-methionine (SAM), and cytoplasmic methyltransferase activities. SAM-dependent methyltransferases methylate a wide range of targets, from small molecules to proteins and nucleic acids. However, few investigations of the global methylome of tumors have been performed. Here, untargeted NMR metabolomics of two mouse tumor models labeled with [13C-methyl]methionine were used to search for the NMR-visible set of cellular methyl acceptors denoted the global methylome. Tumor models were B16 melanoma cell cultures and B16 melanoma tumors, which may be considered as two stages of B16 tumor development. Based on 2D 1H-13C NMR spectra and orthogonal partial least squares discriminant analysis of spectra, our study revealed markedly different global methylomes for melanoma models. The methylome of B16 melanoma cell cultures was dominated by histone methylations, whereas that of B16 melanoma tumors was dominated by cytoplasmic small-molecule methylations. Overall, the technique gave access to the non-DNA methylome. Comparison of tumor models also exhibiting differential expression of aerobic glycolysis provided clues to a methyl metabolism shift during tumor progression.


Assuntos
Melanoma Experimental , Vitamina U , Animais , Epigenoma , Melanoma Experimental/patologia , Metabolômica/métodos , Metionina , Camundongos
2.
Metabolomics ; 14(5): 55, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29937703

RESUMO

INTRODUCTION: Elucidating molecular alterations due to mitochondrial Complex I (CI) mutations may help to understand CI deficiency (CID), not only in mitochondriopathies but also as it is caused by drugs or associated to many diseases. OBJECTIVES: CID metabolic expression was investigated in Leber's hereditary optic neuropathy (LHON) caused by an inherited mutation of CI. METHODS: NMR-based metabolomics analysis was performed in intact skin fibroblasts from LHON patients. It used several datasets: one-dimensional 1H-NMR spectra, two-dimensional 1H-NMR spectra and quantified metabolites. Spectra were analysed using orthogonal partial least squares-discriminant analysis (OPLS-DA), and quantified metabolites using univariate statistics. The response to idebenone (IDE) and resveratrol (RSV), two agents improving CI activity and mitochondrial functions was evaluated. RESULTS: LHON fibroblasts had decreased CI activity (- 43%, p < 0.01). Metabolomics revealed prominent alterations in LHON including the increase of fatty acids (FA), polyunsaturated FA and phosphatidylcholine with a variable importance in the prediction (VIP) > 1 in OPLS-DA, p < 0.01 in univariate statistics, and the decrease of amino acids (AA), predominantly glycine, glutamate, glutamine (VIP > 1) and alanine (VIP > 1, p < 0.05). In LHON, treatment with IDE and RSV increased CI activity (+ 40 and + 44%, p < 0.05). IDE decreased FA, polyunsaturated FA and phosphatidylcholine (p < 0.05), but did not modified AA levels. RSV decreased polyunsaturated FA, and increased several AA (VIP > 1 and/or p < 0.05). CONCLUSION: LHON fibroblasts display lipid and amino acid metabolism alterations that are reversed by mitochondria-targeted treatments, and can be related to adaptive changes. Findings bring insights into molecular changes induced by CI mutation and, beyond, CID of other origins.

3.
Metabolites ; 7(4)2017 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-28937622

RESUMO

There is a rising incidence of non-alcoholic fatty liver disease (NAFLD) as well as of the frequency of Hepato-Cellular Carcinoma (HCC) associated with NAFLD. To seek for putative metabolic pathways specific of the NAFLD etiology, we performed comparative metabolomics between HCC associated with NAFLD and HCC associated with cirrhosis. The study included 28 pairs of HCC tissue versus distant Non-Tumoral Tissue (NTT) collected from patients undergoing hepatectomy. HCC was associated with cirrhosis (n = 9), normal liver (n = 6) and NAFLD (n = 13). Metabolomics was performed using 1H-NMR Spectroscopy on tissue extracts and combined to multivariate statistical analysis. In HCC compared to NTT, statistical models showed high levels of lactate and phosphocholine, and low level of glucose. Shared and Unique Structures (SUS) plots were performed to remove the impact of underlying disease on the metabolic profile of HCC. HCC-cirrhosis was characterized by high levels of ß-hydroxybutyrate, tyrosine, phenylalanine and histidine whereas HCC-NAFLD was characterized by high levels of glutamine/glutamate. In addition, the overexpression glutamine/glutamate on HCC-NAFLD was confirmed by both Glutamine Synthetase (GS) immuno-staining and NMR-spectroscopy glutamine quantification. This study provides evidence of metabolic specificities of HCC associated with non-cirrhotic NAFLD versus HCC associated with cirrhosis. These alterations could suggest activation of glutamine synthetase pathway in HCC-NAFLD and mitochondrial dysfunction in HCC-cirrhosis, that may be part of specific carcinogenic processes.

4.
PLoS One ; 9(7): e102438, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25010005

RESUMO

The hypermetabolic nature of cancer cells and their increased reliance on "aerobic glycolysis", as originally described by Otto Warburg and colleagues, are considered metabolic hallmarks of cancer cells. BRCA1 is a major tumor suppressor in breast cancer and it was implicated in numerous pathways resulting in anticarcinogenic functions. The objective of our study was to address specific contributions of BRCA1 to the metabolic features of cancer cells, including the so-called "Warburg effect". To get a comprehensive approach of the role of BRCA1 in tumor cell metabolism, we performed a global transcriptional and metabolite profiling in a BRCA1-mutated breast cancer cell line transfected or not by wild-type BRCA1. This study revealed that BRCA1 induced numerous modifications of metabolism, including strong inhibition of glycolysis while TCA cycle and oxidative phosphorylation tended to be activated. Regulation of AKT by BRCA1 in both our cell model and BRCA1-mutated breast tumors was suggested to participate in the effect of BRCA1 on glycolysis. We could also show that BRCA1 induced a decrease of ketone bodies and free fatty acids, maybe consumed to supply Acetyl-CoA for TCA cycle. Finally increased activity of antioxidation pathways was observed in BRCA1-transfected cells, that could be a consequence of ROS production by activated oxidative phosphorylation. Our study suggests a new function for BRCA1 in cell metabolic regulation, globally resulting in reversion of the Warburg effect. This could represent a new mechanism by which BRCA1 may exert tumor suppressor function.


Assuntos
Proteína BRCA1/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteínas de Neoplasias/biossíntese , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Ciclo do Ácido Cítrico/genética , Metabolismo Energético/genética , Feminino , Glicólise/genética , Humanos , Mitocôndrias/metabolismo , Fosforilação Oxidativa
5.
Biochim Biophys Acta ; 1840(3): 1092-104, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24296419

RESUMO

BACKGROUND: Localized radiotherapy is long known to cause damages to not only targeted but also non-targeted cells, the so-called bystander (BS) effect. Recently, BS effect was demonstrated in response to chemotherapy. To get further insight into the mechanism of chemotherapy-induced BS effect in vivo, we investigated the response of normal tissues and untreated BS melanomas, at distance from localized chemotherapy-treated melanomas. METHODS: B16 melanoma cells were inoculated sc in one flank, in mice. Chemotherapy was administered intratumorally. After 3 weeks, untreated melanomas were implanted into the other flank. Tumors were analyzed morphologically, and using metabolomics and transcriptomics. RESULTS: Locally-treated melanomas showed growth inhibition and pleiotropic metabolic and transcriptional alterations. Tumors recovered slow proliferation while exhibiting prominent oxidative stress response (decreased glutathione level, and increased expression of genes including Mt1, Gpx3, Sod3, and Hmox1). Plasma contained increased levels of oxidative stress products. However, liver and soleus muscle displayed unaltered morphological characteristics. In contrast, untreated BS melanomas induced from naive B16 cells showed reduced growth, marked oxidative stress response (decreased glutathione level, and increased expression of genes including Sod2, Gpx1 and Gsr), and ras oncogene expression alterations. Furthermore, metabolomics and transcriptomics enabled to estimate the proportion of cells undergoing the BS effect within treated tumors. CONCLUSION: Treatment of tumors with chemotherapy induces BS effects, underpinned by oxidative stress, in abnormal proliferating tissues in vivo, not in normal tissue, that significantly contribute to overall tumor response. General significance BS effect significantly contributes to response to chemotherapy, and may be exploited to improve overall response to cancer treatment.


Assuntos
Antineoplásicos/efeitos adversos , Efeito Espectador , Melanoma Experimental/tratamento farmacológico , Metabolômica , Estresse Oxidativo , Transcriptoma , Trifosfato de Adenosina/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL
6.
Mar Drugs ; 11(10): 3846-60, 2013 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-24152560

RESUMO

Marine natural products are a source of promising agents for cancer treatment. However, there is a need to improve the evaluation of their mechanism of action in tumors. Metabolomics of the response to anti-tumor agents is a tool to reveal candidate biomarkers and metabolic targets. We used two-dimensional high-resolution magic angle spinning proton-NMR spectroscopy-based metabolomics to investigate the response of MCF7 breast cancer cells to ascididemin, a marine alkaloid and lead molecule for anti-cancer treatment. Ascididemin induced severe oxidative stress and apoptosis within 48 h of exposure. Thirty-three metabolites were quantified. Metabolic response involved downregulation of glycolysis and the tricarboxylic acid cycle, and phospholipid metabolism alterations. Candidate metabolic biomarkers of the response of breast cancer cells to ascididemin were proposed including citrate, gluconate, polyunsaturated fatty acids, glycerophospho-choline and -ethanolamine. In addition, candidate metabolic targets were identified. Overall, the response to Asc could be related to severe oxidative stress and anti-inflammatory effects.


Assuntos
Alcaloides/farmacologia , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Metaboloma/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fenantrolinas/farmacologia , Quinolinas/farmacologia , Linhagem Celular Tumoral , Feminino , Humanos , Células MCF-7 , Metabolômica/métodos
7.
PLoS One ; 8(3): e57971, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23472124

RESUMO

BACKGROUND: Curcumin (CUR) has deserved extensive research due to its anti-inflammatory properties, of interest in human diseases including cancer. However, pleiotropic even paradoxical responses of tumor cells have been reported, and the mechanisms of action of CUR remain uncompletely elucidated. METHODOLOGY/PRINCIPAL FINDINGS: (1)H-NMR spectroscopy-based metabolomics was applied to get novel insight into responses of MCF7 and MDA-MB-231 breast cancer cells to CUR alone, and MCF7 cells to CUR in cotreatment with docetaxel (DTX). In both cell types, a major target of CUR was glutathione metabolism. Total glutathione (GSx) increased at low dose CUR (≤ 10 mg.l(-1)-28 µM-) (up to +121% in MCF7 cells, P<0.01, and +138% in MDA-MB-231 cells, P<0.01), but decreased at high dose (≥ 25 mg.l(-1) -70 µM-) (-49%, in MCF7 cells, P<0.02, and -56% in MDA-MB-231 cells, P<0.025). At high dose, in both cell types, GSx-related metabolites decreased, including homocystein, creatine and taurine (-60 to -80%, all, P<0.05). Together with glutathione-S-transferase actvity, data established that GSx biosynthesis was upregulated at low dose, and GSx consumption activated at high dose. Another major target, in both cell types, was lipid metabolism involving, at high doses, accumulation of polyunsaturated and total free fatty acids (between ×4.5 and ×11, P<0.025), and decrease of glycerophospho-ethanolamine and -choline (about -60%, P<0.025). Multivariate statistical analyses showed a metabolic transition, even a biphasic behavior of some metabolites including GSx, between low and high doses. In addition, CUR at 10 mg.l(-1) in cotreatment with DTX induced modifications in glutathione metabolism, lipid metabolism, and glucose utilization. Some of these changes were biphasic depending on the duration of exposure to CUR. CONCLUSIONS/SIGNIFICANCE: Metabolomics reveals major metabolic targets of CUR in breast cancer cells, and biphasic responses that challenge the widely accepted beneficial effects of the phytochemical.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Curcumina/farmacologia , Taxoides/farmacologia , Linhagem Celular Tumoral , Ensaio Cometa , Docetaxel , Feminino , Glutationa/metabolismo , Glutationa Transferase/metabolismo , Humanos , Inflamação , Células MCF-7 , Espectroscopia de Ressonância Magnética , Metabolômica , Análise Multivariada , Estresse Oxidativo , Resultado do Tratamento
8.
Am J Physiol Endocrinol Metab ; 302(11): E1407-18, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22472999

RESUMO

Recent epidemiological studies have suggested a link between cancer and pathophysiological conditions associated with hyperinsulinemia. In this report, we address the possible role of insulin exposure in melanocyte transformation. To this aim, normal melanocytes were exposed to chronic insulin and glucose supplementation (twice the standard medium concentration) for at least 3 wk. After 3-wk treatment, melanocytes increased proliferation (doubling time: 2.7 vs. 5.6 days, P < 0.01). After 3-wk treatment or after 3-wk treatment followed by 4-wk reculture in standard medium, melanocytes were able to grow in soft agar colonies. Treated melanocytes had increased DNA content (+8%, P < 0.05), chromosomal aberrations, and modified oncoprotein profile: p-Akt expression increased (+32%, P < 0.01), Akt decreased, and c-Myc increased (+40%, P < 0.05). PP2A protein expression increased (+42, P < 0.05), while PP2A methylation decreased (-42%, P < 0.05), and PP2A activity was reduced (-27%, P < 0.05). PP2A transcription level was increased (ppp2r1a, PP2A subunit A, +44%, P < 0.05). Also, transcriptomic data revealed modifications in insr (insulin receptors, +10%, P < 0.05) and Il8 (inflammation protein, +99%, P < 0.01). Glycolysis was modified with increased transcription of Pgk1 and Hif1a (P < 0.05), decreased transcription of Pfkfb3 (P < 0.05), decreased activity of pyruvate kinase (P < 0.01), and decreased pyruvate cell content as assessed by (1)H-NMR spectroscopy. In addition, methyl group metabolism was altered with decreased global DNA methylation (-51%, P < 0.01), increased cytosolic protein methylation (+18%, P < 0.05), and consistent changes in methylated species on (1)H-NMR spectra. In conclusion, exposure to chronic insulin and glucose supplementation induces oncogenic changes and methyl group metabolism redistribution, which may be a biomarker of transformation.


Assuntos
Transformação Celular Neoplásica/efeitos dos fármacos , Glucose/farmacologia , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Melanócitos/efeitos dos fármacos , Proteína Fosfatase 2/metabolismo , Western Blotting , Ciclo Celular/efeitos dos fármacos , Fracionamento Celular , Criança , Pré-Escolar , Meios de Cultura , DNA/metabolismo , Relação Dose-Resposta a Droga , Glicólise/efeitos dos fármacos , Humanos , Marcação por Isótopo , Cariotipagem , Espectroscopia de Ressonância Magnética , Masculino , Metilação , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/patologia , Piruvato Quinase/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células-Tronco
9.
Biochim Biophys Acta ; 1810(4): 432-8, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21147198

RESUMO

BACKGROUND: BRCA1, the main breast and ovarian cancer susceptibility gene, has a key role in maintenance of genome stability, cell cycle and transcription regulation. Interestingly, some of the numerous proteins which interact with BRCA1 protein undergo conjugation with small ubiquitin-like modifiers (SUMO). This post-translational modification is related to transcription, DNA repair, nuclear transport, signal transduction, and to cell cycle stress response. METHODS AND RESULTS: Protein sequence analysis suggests that sumoylation target sites belong to the RING finger and BRCT domains (BRCA1 C-terminus), two crucial regions for BRCA1 function. Moreover putative SUMO interacting motifs are present in the sequence of many proteins of BRCA1 network. Using immunoprecipitations and western blotting, we show the conjugation of endogenous nuclear BRCA1 protein with SUMO-2/3. BRCA1 conjugation with SUMO-2/3 is linked to the cell cycle in a cell line dependent manner since no cell cycle dependence of sumoylation is observed in MCF7 breast cancer cells. In contrast, BRCA1 conjugation with SUMO-2/3 is linked to the oxidative stress independently to the cell line, in DU145, MCF7 and 293 T cells. CONCLUSION AND GENERAL SIGNIFICANCE: Our data reveal a new BRCA1 regulation pathway implying sumoylation in response to cell cycle progression and oxidative stress, providing a possible mechanism for the involvement of BRCA1 gene in tumorigenesis.


Assuntos
Proteína BRCA1/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Sequência de Aminoácidos , Animais , Proteína BRCA1/análise , Ciclo Celular , Linhagem Celular , Núcleo Celular/metabolismo , Humanos , Imunoprecipitação , Dados de Sequência Molecular , Estresse Oxidativo , Alinhamento de Sequência , Análise de Sequência de Proteína , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/análise
10.
Biochem Pharmacol ; 80(8): 1170-9, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20637732

RESUMO

Marine plants and animals are sources of a huge number of pharmacologically active compounds, some of which exhibit antineoplastic activity of clinical relevance. However the mechanism of action of marine natural products (MNPs) is poorly understood. In this study, proton NMR spectroscopy-based metabolomics was applied to unravel biochemical disorders induced in human MCF7 breast cancer cells by 3 lead candidate anticancer MNPs: ascididemin (Asc), lamellarin-D (Lam-D), and kahalalide F (KF). Asc, Lam-D, and KF provoked a severe decrease in DNA content in MCF7 cells after 24-h treatment. Asc and Lam-D provoked apoptosis, whereas KF induced non-apoptotic cell death. Metabolite profiling revealed major biochemical disorders following treatment. The response of MCF7 tumor cells to Asc involved the accumulation of citrate (x17 the control level, P<0.001), testifying enzyme blockade in citrate metabolism, and the accumulation of gluconate (x9.8, P<0.005), a metabolite never reported at such concentration in tumor cells, probably testifying glycolysis shutdown. The response to Lam-D involved the accumulation of aspartate (x7.2, P<0.05), glutamate (x14.7, P<0.05), and lactate (x2.3, P<0.05), probably in relation with the targeting of the malate-aspartate shuttle, as discussed. The response to KF involved increased lipid accumulation (polyunsaturated fatty acids x9.8, P<0.05), and phospholipid and acetate derivative alterations. Altogether, this study demonstrates the potential of proton NMR spectroscopy-based metabolomics to help uncover metabolic targets and elucidate the mechanism of cytotoxicity of candidate antineoplastic MNPs.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Espectroscopia de Ressonância Magnética/métodos , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Redes e Vias Metabólicas/efeitos dos fármacos , Metabolômica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
11.
Magn Reson Med ; 63(5): 1172-83, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20432288

RESUMO

NMR spectroscopy-based metabolomics still needs development in quantification procedures. A method was designed for quantitative two-dimensional high resolution magic angle spinning (HRMAS) proton-NMR spectroscopy-based metabolite profiling of intact cells. It uses referencing of metabolite-related NMR signals to protein-related NMR signals and yields straightforward and automatable metabolite profiling. The method enables exploitation of only two-dimensionally visible metabolites and combination of one- and two-dimensional spectra, thus providing an appreciable number of screened metabolites. With this procedure, 32 intracellular metabolites were attributed and quantified in human normal fibroblasts and tumor cells. The phenotype of several tumor cell lines (MCF7, PC3, 143B, and HepG2) was characterized by high levels of glutathione in cell lines with the higher proliferation rate, high levels of creatine, low levels of free amino acids, increased levels of phospholipid derivatives (mostly phosphocholine), and lower lactate content in cell lines with the higher proliferation rate. Other metabolites such as fatty acids differed widely among tumor cell lines. The response of tumor cell lines to chemotherapy also was evaluated by differential metabolite profiling, bringing insights into drug cytotoxicity and tumor cell adaptive mechanisms. The method may prove widely applicable to tumor cell phenotyping.


Assuntos
Antineoplásicos/administração & dosagem , Biomarcadores Tumorais/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Linhagem Celular Tumoral , Humanos , Prótons
12.
J Integr Bioinform ; 7(3)2010 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-20375456

RESUMO

We study, in this paper, a model for the core of the system of the Glycerophospholipid metabolism in the murine cells. It comprises the simple and enzymatic reactions of PhosphatidylEthanolamine and the PhosphatidylCholine. The model's general structure is taken from a number of books and articles. We translate this model into a set of ordinary differential equations (ODEs), to propose a quantitative explanation of the experimental experiences and the observed results. In order to make it usable as a basis for simulations and mathematical analysis we need to make precise the various constants present in the equations but which are usually not directly accessible in the literature. In a first step we considered experimental data of rat's liver cells obtained by NMR spectroscopy: given the values of metabolite concentrations we find appropriate parameter values which allow us to describe the system with ODEs. We have then performed several analyses using the developed model such as stability analysis. A first interesting result is the global stability of the system which was observed by simulation and then proved by mathematical arguments. A second important result is that we observe on the diagrams that the steady state for normal cells is precisely a singular point of order two, whereas tumoral cells present different characteristics; this fact has been proved for PhosphatidylEthanolamine N-Methyl transferase (PEMT), an enzyme which seems to be identified for the first time as a crucial element in the tumoral process. In a second step we applied our model to experimental data of proton HRMAS NMR spectroscopy for solid B16 melanoma and Lewis lung (3LL) 3LL carcinoma cells treated by Chloroethyl Nitrosourea (CENU). We performed a complete comparative analysis of parameters in order to learn the predictive statements to explain increases and decreases which one can observe in concentrations.


Assuntos
Antineoplásicos/uso terapêutico , Etilnitrosoureia/análogos & derivados , Modelos Biológicos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Fosfolipídeos/biossíntese , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Etilnitrosoureia/farmacologia , Etilnitrosoureia/uso terapêutico , Cinética , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilcolinas/metabolismo , Fosfatidiletanolaminas/metabolismo
13.
Breast Cancer Res Treat ; 120(3): 613-26, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19513827

RESUMO

There is growing evidence that docetaxel, a microtubule-targeting agent like the other taxane paclitaxel, induces dual cytotoxicity mechanism according to dose level. Postgenomics screening technologies are now more and more applied to the elucidation of drug response mechanisms. Proton nuclear magnetic resonance spectroscopy-based pharmacometabolomics was here applied to get further insight into the response of human MCF7 breast carcinoma cells to docetaxel at high (clinical, 5 microM) and low (1 nM) doses. The global response to both doses was evaluated by nuclear morphology and DNA content, the latter as an index of cell proliferation and DNA ploidy. High dose provoked long-lasting cell cycle arrest in mitosis during the first 48 h of exposure to treatment and severe decrease in DNA content followed by significant amount of cell death. In contrast, at low dose, no long-lasting cell cycle arrest was observed on micrographies, and DNA content was decreased but less than at high dose (P < 0.05), without significant cell death. This response was compared to biochemical alteration assessed by pharmacometabolomics. Thirty metabolites were identified and quantified. Metabolite profiling at clinical dose revealed time-dependent disorders in derivatives of glycolysis, lipid metabolism and glutathione metabolism. Comparison between high and low doses was performed at 72 h and showed common traits including the accumulation of cytidinediphosphocholine (x 5.0 and x 6.9, respectively, P < 0.03), the decrease in phosphatidylcholine (x 0.3 and x 0.2, respectively, P < 0.03), and gluthathione (x 0.6 and x 0.6, respectively, P < 0.03). Despite that, significant dose-dependent differences were found in 12 of 30 measured metabolites. Among them, the most discriminant metabolites were polyunsaturated fatty acids (ratio of high-to-low dose of 14.8, P < 0.05), glutamate, myoinositol, and homocysteine (ratio < 0.4, P < 0.05). In addition, the mechanism for glutathione decrease was different. At high dose, it resulted from extensive consumption with precursor starvation (glutamate: -89%, P < 0.05) and increased glutathione S-transferase activity (x 5, P < 0.01), whereas at low dose, it resulted from glutathione biosynthesis blockade with homocysteine accumulation (+144%, P < 0.03) and decreased glutathione S-transferase activity (-70%, P < 0.01). Altogether, this pharmacometabolomics analysis provides further evidence of the varying cellular responses at high and low doses of docetaxel in MCF7 breast cancer cells.


Assuntos
Adenocarcinoma/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/metabolismo , Redes e Vias Metabólicas/efeitos dos fármacos , Taxoides/farmacologia , Adenocarcinoma/patologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/ultraestrutura , DNA de Neoplasias/análise , Docetaxel , Relação Dose-Resposta a Droga , Feminino , Glutationa/metabolismo , Humanos , Metabolômica , Mitose/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Ressonância Magnética Nuclear Biomolecular , Fosfolipídeos/metabolismo
14.
Nutr Cancer ; 61(4): 518-29, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19838924

RESUMO

Methionine (Met) deprivation stress (MDS) is proposed in association with chemotherapy in the treatment of some cancers. A synergistic effect of this combination is generally acknowledged. However, little is known on the mechanism of the response to this therapeutic strategy. A model of B16 melanoma tumor in vivo was treated by MDS alone and in combination with chloroethylnitrosourea (CENU). It was applied recent developments in proton-NMR spectroscopy-based metabolomics for providing information on the metabolic response of tumors to MDS and combination with chemotherapy. MDS inhibited tumor growth during the deprivation period and growth resumption thereafter. The combination of MDS with CENU induced an effective time-dependent synergy on growth inhibition. Metabolite profiling during MDS showed a decreased Met content (P < 0.01) despite the preservation of the protein content, disorders in sulfur-containing amino acids, glutamine/proline, and phospholipid metabolism [increase of glycerophosphorylcholine (P < 0.01), decrease in phosphocholine (P < 0.05)]. The metabolic profile of MDS combined with CENU and ANOVA analysis revealed the implication of Met and phospholipid metabolism in the observed synergy, which may be interpreted as a Met-sparing metabolic reprogramming of tumors. It follows that combination therapy of MDS with CENU seems to intensify adaptive processes, which may set limitations to this therapeutic strategy.


Assuntos
Adaptação Fisiológica/efeitos dos fármacos , Antineoplásicos/uso terapêutico , Melanoma Experimental/terapia , Metionina/deficiência , Metionina/metabolismo , Compostos de Nitrosoureia/uso terapêutico , Fosfolipídeos/metabolismo , Análise de Variância , Animais , Peso Corporal/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Terapia Combinada , Intervalos de Confiança , Inibidores do Crescimento/uso terapêutico , Fígado/efeitos dos fármacos , Masculino , Melanoma Experimental/química , Melanoma Experimental/dietoterapia , Melanoma Experimental/metabolismo , Metaboloma/efeitos dos fármacos , Metionina/análise , Camundongos , Camundongos Endogâmicos C57BL , Ressonância Magnética Nuclear Biomolecular , Tamanho do Órgão/efeitos dos fármacos , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Fosfolipídeos/análise , Distribuição Aleatória , Estresse Fisiológico/efeitos dos fármacos , Fatores de Tempo , Carga Tumoral/efeitos dos fármacos
15.
Mol Carcinog ; 48(8): 733-41, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19347860

RESUMO

Cancer cells mainly rely on glycolysis for energetic needs, and mitochondrial ATP production is almost inactive. However, cancer cells require the integrity of mitochondrial functions for their survival, such as the maintenance of the internal membrane potential gradient (DeltaPsim). It thus may be predicted that DeltaPsim regeneration should depend on cellular capability to produce sufficient ATP by upregulating glycolysis or recruiting oxidative phosphorylation (OXPHOS). To investigate this hypothesis, we compared the response to an anticancer agent chloroethylnitrosourea (CENU) of two transformed cell lines: HepG2 (hepatocarcinoma) with a partially differentiated phenotype and 143B (osteosarcoma) with an undifferentiated one. These cells types differ by their mitochondrial OXPHOS background; the most severely impaired being that of 143B cells. Treatment effects were tested on cell proliferation, O(2) consumption/ATP production coupling, DeltaPsim maintenance, and global metabolite profiling by NMR spectroscopy. Our results showed an OXPHOS uncoupling and a lowered DeltaPsim, leading to an increased energy request to regenerate DeltaPsim in both models. However, energy request could not be met by undifferentiated cells 143B, which ATP content decreased after 48 h leading to cell death, while partially differentiated cells (HepG2) could activate their oxidative metabolism and escape chemotherapy. We propose that mitochondrial OXPHOS background confers a survival advantage to more differentiated cells in response to chemotherapy. This suggests that the mitochondrial bioenergetic background of tumors should be considered for anticancer treatment personalization.


Assuntos
Carcinoma Hepatocelular/metabolismo , Metabolismo Energético , Mitocôndrias/metabolismo , Osteossarcoma/metabolismo , Trifosfato de Adenosina/metabolismo , Carcinoma Hepatocelular/tratamento farmacológico , Diferenciação Celular , Proliferação de Células , Respiração Celular , Sobrevivência Celular/efeitos dos fármacos , Etilnitrosoureia/análogos & derivados , Etilnitrosoureia/farmacologia , Glucose/metabolismo , Humanos , Ácido Láctico/metabolismo , Espectroscopia de Ressonância Magnética , Potencial da Membrana Mitocondrial , Mitocôndrias/efeitos dos fármacos , Osteossarcoma/tratamento farmacológico , Fosforilação Oxidativa , Ácido Pirúvico/metabolismo , Células Tumorais Cultivadas
16.
Anticancer Res ; 28(1A): 21-7, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18383820

RESUMO

Chloroethylnitrosourea (CENU) chemotherapy is used for the treatment of melanoma tumors. The main mechanism of action of this anticancer agent is via DNA damage. We recently showed in murine experiments using a parental double B16 melanoma tumor model that, after treatment of primary tumors with cystemustine (CENU agent), untreated secondary tumors exhibited growth inhibition and metabolism disorders. The response of secondary untreated tumor was called the chemotherapy-induced bystander effect. To see whether chemotherapy-induced bystander effects were induced with other members of the CENU family, we compared three CENU(s) used in melanoma treatment: cystemustine, carmustine and fotemustine. Our results demonstrate that fotemustine, like cystemustine, but not carmustine induced a protective effect against secondary untreated tumors including alterations in phospholipid derivative and glutathione which are the metabolic signature of the bystander effect. From these data we may conclude that DNA damage to the primary tumor is not sufficient to explain chemotherapy-induced bystander effects.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Dano ao DNA , Melanoma Experimental/tratamento farmacológico , Compostos de Nitrosoureia/farmacologia , Cloreto de Vinil/análogos & derivados , Animais , Carmustina/farmacologia , Glutationa/metabolismo , Masculino , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Segunda Neoplasia Primária/prevenção & controle , Ressonância Magnética Nuclear Biomolecular , Compostos Organofosforados/farmacologia , Fosfolipídeos/metabolismo
17.
Int J Oncol ; 32(1): 49-57, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18097542

RESUMO

Protein phosphatase 2A (PP2A), an Akt pathway inhibitor, is considered to be activated by methylation of its catalytic subunit. Also PP2A downregulation was proposed to take part in carcinogenesis. Recently, PP2A activation was shown to be activated in response to DNA damage. To obtain further information on the role of PP2A in tumors and response to DNA damage, we investigated the relationship between PP2A methylation and activity, cell proliferation, Akt activation, c-Myc expression and PTEN activity in B16 melanoma cells untreated and after chloroethylnitrosourea (CENU) treatment. In untreated cells, okadaic acid, an antagonist of PP2A methylation, inhibited PP2A activity, stimulated cell proliferation, increased Akt activation and c-Myc expression. Xylulose-5-phosphate, an agonist of PP2A methylation, increased PP2A activity, decreased cell proliferation, Akt activation and c-Myc expression. However, both PP2A methylation modulators increased PTEN activity. During the response to CENU treatment, PP2A methylation and activity were strongly increased, Akt activation and c-Myc expression were decreased. However PTEN activity was increased. After tumor cell growth recovery, these modifications were moderately decreased. PP2A methylation was quantified and correlated positively with PP2A activity, and negatively with criteria for cell aggressiveness (cell proliferation, Akt activation, c-Myc expression). Based on these data, PP2A methylation status controls PP2A activity and oncoproteins expression and PP2A is strongly activated after CENU treatment thus partly explaining the growth inhibition in response to this agent. It follows that PP2A promethylating agents are potential candidates for anticancer drugs.


Assuntos
Antineoplásicos/farmacologia , Melanoma Experimental/tratamento farmacológico , Compostos de Nitrosoureia/farmacologia , Proteína Fosfatase 2/metabolismo , Animais , Dano ao DNA , Melanoma Experimental/patologia , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Ácido Okadáico/farmacologia , PTEN Fosfo-Hidrolase/metabolismo , Pentosefosfatos/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo
18.
Cancer Res ; 67(5): 2150-9, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17332345

RESUMO

Metabolomics of tumors may allow discovery of tumor biomarkers and metabolic therapeutic targets. Metabolomics by two-dimensional proton high-resolution magic angle spinning nuclear magnetic resonance spectroscopy was applied to investigate metabolite disorders following treatment by chloroethylnitrosourea of murine B16 melanoma (n = 33) and 3LL pulmonary carcinoma (n = 31) in vivo. Treated tumors of both types resumed growth after a delay. Nitrosoureas provoke DNA damage but the metabolic consequences of genotoxic stress are little known yet. Although some differences were observed in the metabolite profile of untreated tumor types, the prominent metabolic features of the response to nitrosourea were common to both. During the growth inhibition phase, there was an accumulation of glucose (more than x10; P < 0.05), glutamine (x3 to 4; P < 0.01), and aspartate (x2 to 5; P < 0.01). This response testified to nucleoside de novo synthesis down-regulation and drug efficacy. However, this phase also involved the increase in alanine (P < 0.001 in B16 melanoma), the decrease in succinate (P < 0.001), and the accumulation of serine-derived metabolites (glycine, phosphoethanolamine, and formate; P < 0.01). This response witnessed the activation of pathways implicated in energy production and resumption of nucleotide de novo synthesis, thus metabolic pathways of DNA repair and adaptation to treatment. During the growth recovery phase, it remained polyunsaturated fatty acid accumulation (x1.5 to 2; P < 0.05) and reduced utilization of glucose compared with glutamine (P < 0.05), a metabolic fingerprint of adaptation. Thus, this study provides the proof of principle that metabolomics of tumor response to an anticancer agent may help discover metabolic pathways of drug efficacy and adaptation to treatment.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Espectroscopia de Ressonância Magnética/métodos , Redes e Vias Metabólicas , Compostos de Nitrosoureia/farmacocinética , Proteômica/métodos , Animais , Antineoplásicos/farmacocinética , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Inativação Metabólica , Masculino , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Transplante de Neoplasias , Compostos de Nitrosoureia/farmacologia , Resultado do Tratamento , Células Tumorais Cultivadas
19.
Int J Oncol ; 29(3): 637-42, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16865279

RESUMO

We recently showed, using a parental double B16 melanoma tumor model that, in the presence of CENU-treated primary tumors, untreated secondary tumors exhibited growth inhibition. This response was shown to be related to CENU-induced bystander effects. To see whether CENU-induced bystander effects were still effective on non-parental syngeneic secondary tumors, Lewis lung (3LL) secondary tumors were inoculated in recipients bearing CENU-treated B16 melanoma tumors. Our results show that non-parental secondary 3LL tumors underwent growth inhibition, differentiation, and phospholipid metabolism alterations, all changes similar to those of parental secondary 3LL tumors. This demonstrates the lack of tumor tissue specificity of chemotherapy-induced bystander effects.


Assuntos
Efeito Espectador/efeitos dos fármacos , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Etilnitrosoureia/análogos & derivados , Neoplasias Pulmonares/tratamento farmacológico , Melanoma Experimental/tratamento farmacológico , Compostos Nitrosos/farmacologia , Fosfolipídeos/metabolismo , Animais , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Diferenciação Celular , Etilnitrosoureia/uso terapêutico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Espectroscopia de Ressonância Magnética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Prótons
20.
Magn Reson Med ; 55(5): 984-96, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16598721

RESUMO

Tumors frequently have abnormal L-methionine (Met) metabolism, the so-called Met-dependence phenotype that refers to the inability to proliferate in the absence of Met. However, the origin of this phenotype is still unknown and may arise from one of several pathways of Met metabolism. To help characterize the metabolic features of Met-dependent/independent phenotypes, the fate of the methyl carbon of L-[methyl-13C]Met was chased in a murine model of malignant melanoma (B16-F1) in vitro and in vivo. Growth curves under Met restriction showed that melanoma cells in vitro were Met-independent, whereas implanted melanoma tumors in vivo were Met-dependent. Label-assisted high-resolution magic angle spinning 1H-13C NMR spectroscopy metabolite profiling showed that, in vitro, creatine and phosphatidylcholine 13C-enrichments were poor, but S-adenosyl-Met and posttranslationally N-methylated protein signals were strong. In contrast, in vivo, creatine and phosphatidylcholine enrichments were strong but S-adenosyl-Met and N-methylated protein signals were poor. In addition, in vivo, transsulfuration was very efficient, consumed one-carbon units originating from the methyl carbon of Met, and yielded taurine labeling. From these data, the Met-dependent/independent phenotypes appear closely related to the source of one-carbon units. Thus, L-[methyl-13C]Met-assisted NMR spectroscopy metabolite profiling allowed the discrimination between Met-dependence and Met-independence and provided novel mechanistic information on their origin.


Assuntos
Biomarcadores Tumorais/análise , Diagnóstico por Computador/métodos , Espectroscopia de Ressonância Magnética/métodos , Melanoma Experimental/classificação , Melanoma Experimental/metabolismo , Metionina/análise , Algoritmos , Animais , Isótopos de Carbono , Masculino , Melanoma Experimental/diagnóstico , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Prótons , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Marcadores de Spin
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA