Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 4: e877, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24157871

RESUMEN

Recent observations on cancer cell metabolism indicate increased serine synthesis from glucose as a marker of poor prognosis. We have predicted that a fraction of the synthesized serine is routed to a pathway for ATP production. The pathway is composed by reactions from serine synthesis, one-carbon (folate) metabolism and the glycine cleavage system (SOG pathway). Here we show that the SOG pathway is upregulated at the level of gene expression in a subset of human tumors and that its level of expression correlates with gene signatures of cell proliferation and Myc target activation. We have also estimated the SOG pathway metabolic flux in the NCI60 tumor-derived cell lines, using previously reported exchange fluxes and a personalized model of cell metabolism. We find that the estimated rates of reactions in the SOG pathway are highly correlated with the proliferation rates of these cell lines. We also observe that the SOG pathway contributes significantly to the energy requirements of biosynthesis, to the NADPH requirement for fatty acid synthesis and to the synthesis of purines. Finally, when the PC-3 prostate cancer cell line is treated with the antifolate methotrexate, we observe a decrease in the ATP levels, AMP kinase activation and a decrease in ribonucleotides and fatty acids synthesized from [1,2-(13)C2]-D-glucose as the single tracer. Taken together our results indicate that the SOG pathway activity increases with the rate of cell proliferation and it contributes to the biosynthetic requirements of purines, ATP and NADPH of cancer cells.


Asunto(s)
Adenosina Trifosfato/metabolismo , Ácido Fólico/metabolismo , Glicina/metabolismo , NADP/metabolismo , Neoplasias/metabolismo , Purinas/metabolismo , Serina/metabolismo , Aminoácido Oxidorreductasas/genética , Animales , Proteínas Portadoras/genética , Línea Celular Tumoral , Células Madre Embrionarias/metabolismo , Metabolismo Energético/efectos de los fármacos , Ácidos Grasos/biosíntesis , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Análisis de Flujos Metabólicos , Redes y Vías Metabólicas , Metotrexato/farmacología , Ratones , Complejos Multienzimáticos/genética , Neoplasias/genética , Biosíntesis de Proteínas , Transferasas/genética
2.
Ernst Schering Found Symp Proc ; (4): 189-203, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18811058

RESUMEN

The metabolic phenotype of tumor cells promote the proliferative state, which indicates that (a) cell transformation is associated with the activation of specific metabolic substrate channels toward nucleic acid synthesis and (b) increased expression phosphorylation, allosteric or transcriptional regulation of intermediary metabolic enzymes and their substrate availability together mediate unlimited growth. It is evident that cell transformation due to various K-ras point mutations is associated with the activation of specific metabolic substrate channels that increase glucose channeling toward nucleic acid synthesis. Therefore, phosphorylation, allosteric and transcriptional regulation of intermediary metabolic enzymes and their substrate availability together mediate cell transformation and growth. In this review, we summarize opposite changes in metabolic phenotypes induced by various cell-transforming agents, and tumor growth-inhibiting drugs or phytochemicals, or novel synthetic antileukemic drugs such as imatinib mesylate (Gleevec). Metabolic enzymes that further incite growth signaling pathways and thus promote malignant cell transformation serve as high-efficacy nongenetic novel targets for cancer therapies.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Humanos , Neoplasias/patología , Oxidación-Reducción , Fenotipo , Transducción de Señal
3.
Br J Cancer ; 91(12): 2094-100, 2004 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-15599384

RESUMEN

Inhibitors of glycogen breakdown regulate glucose homeostasis by limiting glucose production in diabetes. Here we demonstrate that restrained glycogen breakdown also inhibits cancer cell proliferation and induces apoptosis through limiting glucose oxidation, as well as nucleic acid and de novo fatty acid synthesis. Increasing doses (50-100 microM) of the glycogen phosphorylase inhibitor CP-320626 inhibited [1,2-(13)C(2)]glucose stable isotope substrate re-distribution among glycolysis, pentose and de novo fatty acid synthesis in MIA pancreatic adenocarcinoma cells. Limited oxidative pentose-phosphate synthesis, glucose contribution to acetyl CoA and de novo fatty acid synthesis closely correlated with decreased cell proliferation. The stable isotope-based dynamic metabolic profile of MIA cells indicated a significant dose-dependent decrease in macromolecule synthesis, which was detected at lower drug doses and before the appearance of apoptosis markers. Normal fibroblasts (CRL-1501) did not show morphological or metabolic signs of apoptosis likely due to their slow rate of growth and metabolic activity. This indicates that limiting carbon re-cycling and rapid substrate mobilisation from glycogen may be an effective and selective target site for new drug development in rapidly dividing cancer cells. In conclusion, pancreatic cancer cell growth arrest and death are closely associated with a characteristic decrease in glycogen breakdown and glucose carbon re-distribution towards RNA/DNA and fatty acids during CP-320626 treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Amidas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Glucógeno Fosforilasa/antagonistas & inhibidores , Glucólisis/efectos de los fármacos , Humanos , Etiquetado Corte-Fin in Situ , Indoles
4.
J Mol Endocrinol ; 29(3): 347-60, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12459036

RESUMEN

The intestinal hormone glucagon-like peptide-1 (GLP-1) has been shown to promote an increase in pancreatic beta-cell mass via proliferation of islet cells and differentiation of non-insulin-secreting cells. In this study, we have characterized some of the events that lead to the differentiation of pancreatic ductal cells in response to treatment with human GLP-1. Rat pancreatic ductal (ARIP) cells were cultured in the presence of GLP-1 and analyzed for cell counting, cell cycle distribution, expression of cyclin-dependent-kinase (Cdk) inhibitors, transcription of beta-cell-specific genes, loss of ductal-like phenotype and acquisition of beta-cell-like gene expression profile. Exposure of ARIP cells to 10 nM GLP-1 induced a significant reduction in the cell replication rate and a significant decrease in the percentage of cells in S phase of the cell cycle. This was associated with an increase in the number of cells in G0-G1 phase and a reduction of cells in G2-M phase. Western blot analysis for the Cdk inhibitors, kinase inhibitor protein 1 (p27(Kip1)) and Cdk-interacting protein 1 (p21(Cip1)), demonstrated a significant increase in p27(Kip1) and p21(Cip1) levels within the first 24 h from the beginning of GLP-1 treatment. As cells slowed down their proliferation rate, GLP-1 also induced a time-dependent expression of various beta-cell-specific mRNAs. The glucose transporter GLUT-2 was the first of those factors to be expressed (24 h treatment), followed by insulin (44 h) and finally by the enzyme glucokinase (56 h). In addition, immunocytochemistry analysis showed that GLP-1 induced a time-dependent down-regulation of the ductal marker cytokeratin-20 (CK-20) and a time-dependent induction of insulin expression. Finally, GLP-1 promoted a glucose-dependent secretion of insulin, as demonstrated by HPLC and RIA analyses of the cell culture medium. The present study has demonstrated that GLP-1 induces a cell cycle re-distribution with a decrease in cell proliferation rate prior to promoting the differentiation of cells towards an endocrine-like phenotype.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Glucagón/farmacología , Conductos Pancreáticos/citología , Conductos Pancreáticos/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Precursores de Proteínas/farmacología , Animales , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Péptido 1 Similar al Glucagón , Glucoquinasa/metabolismo , Glucosa/farmacología , Transportador de Glucosa de Tipo 2 , Humanos , Inmunohistoquímica , Insulina/análisis , Insulina/metabolismo , Secreción de Insulina , Proteínas de Transporte de Monosacáridos/metabolismo , Ratas , Proteínas Supresoras de Tumor/metabolismo
5.
Leuk Res ; 25(10): 883-90, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11532522

RESUMEN

The organophosphate pesticide, isofenphos, is associated with human myeloid leukemia. In this study we describe metabolic changes in K562 myeloid blast cells from exposure to varying concentrations of isofenphos using the stable [1,2-13C(2)]glucose isotope as the single tracer and biological mass spectrometry. Isofenphos (1, 10, 100 microg/ml/72 h) treated K562 cells showed increases of 10.7, 33.8 and 39.7% in lactate production as well as a 14.2% increase (1 microg/ml/72 h) in 13C incorporation into nucleic acid ribose from glucose. Concomitantly, we observed a decrease in glucose oxidation and the synthesis of glutamate, palmitate and stearate from glucose. Our results demonstrate that this organophosphate pesticide exerts a leukemogenic effect by the recruitment of glucose carbons for nucleic acid synthesis thus promoting proliferation simultaneous with poor differentiation. The imbalanced metabolic phenotype with a severe defect in glucose oxidation, lipid and amino acid synthesis concurrent with de novo synthesis of nucleic acids in response to isofenphos treatment conforms to the invasive proliferating phenotype observed in TGF-beta treated lung epithelial carcinoma cells.


Asunto(s)
Insecticidas/farmacología , Células K562/efectos de los fármacos , Compuestos Organotiofosforados/farmacología , Ácidos Grasos/análisis , Glucosa/metabolismo , Ácido Glutámico/metabolismo , Humanos , Células K562/metabolismo , Ácido Láctico/metabolismo , Espectrometría de Masas , Vía de Pentosa Fosfato , ARN/metabolismo , Ribosa/metabolismo
6.
Eur J Biochem ; 268(15): 4177-82, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11488910

RESUMEN

Thiamine deficiency frequently occurs in patients with advanced cancer and therefore thiamine supplementation is used as nutritional support. Thiamine (vitamin B1) is metabolized to thiamine pyrophosphate, the cofactor of transketolase, which is involved in ribose synthesis, necessary for cell replication. Thus, it is important to determine whether the benefits of thiamine supplementation outweigh the risks of tumor proliferation. Using oxythiamine (an irreversible inhibitor of transketolase) and metabolic control analysis (MCA) methods, we measured an in vivo tumour growth control coefficient of 0.9 for the thiamine-transketolase complex in mice with Ehrlich's ascites tumour. Thus, transketolase enzyme and thiamine clearly determine cell proliferation in the Ehrlich's ascites tumour model. This high control coefficient allows us to predict that in advanced tumours, which are commonly thiamine deficient, supplementation of thiamine could significantly increase tumour growth through transketolase activation. The effect of thiamine supplementation on tumour proliferation was demonstrated by in vivo experiments in mice with the ascites tumour. Thiamine supplementation in doses between 12.5 and 250 times the recommended dietary allowance (RDA) for mice were administered starting on day four of tumour inoculation. We observed a high stimulatory effect on tumour growth of 164% compared to controls at a thiamine dose of 25 times the RDA. This growth stimulatory effect was predicted on the basis of correction of the pre-existing level of thiamine deficiency (42%), as assayed by the cofactor/enzyme ratio. Interestingly, at very high overdoses of thiamine, approximately 2500 times the RDA, thiamine supplementation had the opposite effect and caused 10% inhibition of tumour growth. This effect was heightened, resulting in a 36% decrease, when thiamine supplementation was administered from the 7th day prior to tumour inoculation. Our results show that thiamine supplementation sufficient to correct existing thiamine deficiency stimulates tumour proliferation as predicted by MCA. The tumour inhibitory effect at high doses of thiamine is unexplained and merits further study.


Asunto(s)
Suplementos Dietéticos , Neoplasias/tratamiento farmacológico , Tiamina/uso terapéutico , Animales , Carcinoma de Ehrlich/tratamiento farmacológico , Carcinoma de Ehrlich/prevención & control , División Celular , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Glucosa-6-Fosfato/farmacología , Ratones , Ratones Endogámicos C57BL , Neoplasias/prevención & control , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/prevención & control , Oxitiamina/farmacología , Tiamina/química , Factores de Tiempo , Transcetolasa/metabolismo , Transcetolasa/farmacología
7.
J Biol Chem ; 276(41): 37747-53, 2001 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-11489902

RESUMEN

Chronic myeloid leukemia cells contain a constitutively active Bcr-Abl tyrosine kinase, the target protein of Gleevec (STI571) phenylaminopyrimidine class protein kinase inhibitor. Here we provide evidence for metabolic phenotypic changes in cultured K562 human myeloid blast cells after treatment with increasing doses of STI571 using [1,2-13C2]glucose as the single tracer and biological mass spectrometry. In response to 0.68 and 6.8 microm STI571, proliferation of Bcr-Abl-positive K562 cells showed a 57% and 74% decrease, respectively, whereas glucose label incorporation into RNA decreased by 13.4% and 30.1%, respectively, through direct glucose oxidation, as indicated by the decrease in the m1/Sigma(m)n ratio in RNA. Based on the in vitro proliferation data, the IC50 of STI571 in K562 cultures is 0.56 microm. The decrease in 13C label incorporation into RNA ribose was accompanied by a significant fall in hexokinase and glucose-6-phosphate 1-dehydrogenase activities. The activity of transketolase, the enzyme responsible for nonoxidative ribose synthesis in the pentose cycle, was less affected, and there was a relative increase in glucose carbon incorporation into RNA through nonoxidative synthesis as indicated by the increase in the m2/Sigma(m)n ratio in RNA. The restricted use of glucose carbons for de novo nucleic acid and fatty acid synthesis by altering metabolic enzyme activities and pathway carbon flux of the pentose cycle constitutes the underlying mechanism by which STI571 inhibits leukemia cell glucose substrate utilization and growth. The administration of specific hexokinase/glucose-6-phosphate 1-dehydrogenase inhibitor anti-metabolite substrates or competitive enzyme inhibitor compounds, alone or in combination, should be explored for the treatment of STI571-resistant advanced leukemias as well as that of Bcr-Abl-negative human malignancies.


Asunto(s)
Ácidos Grasos/biosíntesis , Glucosa/metabolismo , Glucosafosfato Deshidrogenasa/metabolismo , Hexoquinasa/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Ácidos Nucleicos/biosíntesis , Piperazinas/farmacología , Pirimidinas/farmacología , Transcetolasa/metabolismo , Antineoplásicos/farmacología , Benzamidas , Carbono/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Células Tumorales Cultivadas
8.
Pancreas ; 23(2): 141-7, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11484916

RESUMEN

The fermented wheat germ extract with standardized benzoquinone composition has potent tumor propagation inhibitory properties. The authors show that this extract induces profound metabolic changes in cultured MIA pancreatic adenocarcinoma cells when the [1,2-13C2]glucose isotope is used as the single tracer with biologic gas chromatography-mass spectrometry. MIA cells treated with 0.1, 1, and 10 mg/mL wheat germ extract showed a dose-dependent decrease in cell glucose consumption. uptake of isotope into ribosomal RNA (2.4%, 9.4%, and 28.0%), and release of 13CO2. Conversely, direct glucose oxidation and ribose recycling in the pentose cycle showed a dose-dependent increase of 1.2%, 20.7%, and 93.4%. The newly synthesized fraction of cell palmitate and the 13C enrichment of acetyl units were also significantly increased with all doses of wheat germ extract. The fermented wheat germ extract controls tumor propagation primarily by regulating glucose carbon redistribution between cell proliferation-related and cell differentiation-related macromolecules. Wheat germ extract treatment is likely associated with the phosphorylation and transcriptional regulation of metabolic enzymes that are involved in glucose carbon redistribution between cell proliferation-related structural and functional macromolecules (RNA, DNA) and the direct oxidative degradation of glucose, which have devastating consequences for the proliferation and survival of pancreatic adenocarcinoma cells in culture.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Extractos Vegetales/farmacología , Ácidos Grasos/biosíntesis , Fermentación , Cromatografía de Gases y Espectrometría de Masas , Glucosa/metabolismo , Humanos , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , ARN Ribosómico/biosíntesis , Ribosa/biosíntesis , Triticum , Células Tumorales Cultivadas
9.
Pancreas ; 22(1): 1-7, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11138960

RESUMEN

Genistein is a plant isoflavonoid bearing potent tumor growth-regulating characteristics. This effect of genistein has been attributed partially to its tyrosine kinase-regulating properties, resulting in cell-cycle arrest and limited angiogenesis. Genistein has been used in chemotherapy-resistant cases of advanced leukemia with promising results. Here we demonstrate that genistein primarily affects nucleic acid synthesis and glucose oxidation in tumor cells using the [1,2-(13)C2]glucose isotope as the single tracer and gas chromatography/mass spectrometry to follow various intracellular glucose metabolites. The ribose fraction of RNA demonstrated a rapid 4.6%, 16.4%, and 46.3% decrease in isotope uptake through the nonoxidative branch of the pentose cycle and a sharp 4.8%. 24.6%, and 48% decrease in 13CO2 release from glucose after 2, 20, and 200 micromol/L genistein treatment, respectively. Fatty acid synthesis and the 13C enrichment of acetyl units were not significantly affected by genistein treatment. De novo glycogen synthesis from media glucose was not detected in cultured MIA cells. It can be concluded from these studies that genistein controls tumor growth primarily through the regulation of glucose metabolism, specifically targeting glucose carbon incorporation into nucleic acid ribose through the nonoxidative steps of the pentose cycle, which represents a new paradigm for the antiproliferative action of a plant phytochemical.


Asunto(s)
Adenocarcinoma/prevención & control , Antineoplásicos/farmacología , Genisteína/farmacología , Neoplasias Pancreáticas/prevención & control , Vía de Pentosa Fosfato/efectos de los fármacos , Ribosa/biosíntesis , Adenocarcinoma/metabolismo , Ciclo Celular/efectos de los fármacos , Cromatografía de Gases y Espectrometría de Masas , Glucosa/metabolismo , Humanos , Neoplasias Pancreáticas/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
10.
Anticancer Res ; 20(3B): 2245-8, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10928186

RESUMEN

The role of food supplements in the form of vitamins has not been extensively investigated in relation to varying cancer rates between populations of different geographical regions. New data indicate that thiamine (vitamin B1), a common food supplement in Western food products, is directly involved in nucleic acid ribose synthesis of tumor cells in its biologically activated form through the non-oxidative transketolase catalyzed pentose cycle reaction. Whether thiamine plays a role in increased cancer rates in the Western World by enhancing tumor cell proliferation, while increased consumption of thiaminase rich food limiting thiamine availability protects against common malignancies in Asia and Africa has not been evaluated. In the Western World, thiamine is a popular vitamin supplement in the form of tablets and it is also added to basic food items such as milled flour, cereals, peanut butter, refreshment drinks and pastas. On the contrary, thiaminase, the natural thiamine-degrading enzyme, is abundantly present in raw and fermented fish, certain vegetables and roasted insects consumed primarily in Africa and Asia. Excess thiamine supplementation in common food products may contribute to the increased cancer rates of the Western World.


Asunto(s)
Suplementos Dietéticos/efectos adversos , Neoplasias/epidemiología , Tiamina/efectos adversos , África/epidemiología , Animales , Asia/epidemiología , ADN de Neoplasias/biosíntesis , Encuestas sobre Dietas , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/metabolismo , Proteínas en la Dieta/metabolismo , Ingestión de Energía , Inducción Enzimática , Europa (Continente)/epidemiología , Peces , Humanos , Hidrolasas/metabolismo , Carne , Neoplasias/inducido químicamente , América del Norte/epidemiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Vía de Pentosa Fosfato , ARN Neoplásico/biosíntesis , Deficiencia de Tiamina/epidemiología , Tiamina Pirofosfato/metabolismo , Transcetolasa/biosíntesis
11.
Am J Physiol Endocrinol Metab ; 279(2): E425-32, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10913044

RESUMEN

We present here a study on the role of leptin in the regulation of lipogenesis by examining the effect of dietary macronutrient composition on lipogenesis in the leptin receptor-defective Zucker diabetic fatty rat (ZDF) and its lean litter mate (ZL). Animals were pair fed two isocaloric diets differing in their fat-to-carbohydrate ratio providing 10 and 30% energy as fat. Lipogenesis was measured in the rats using deuterated water and isotopomer analysis. From the deuterium incorporation into plasma palmitate, stearate, and oleate, we determined de novo synthesis of palmitate and synthesis of stearate by chain elongation and of oleate by desaturation. Because the macronutrient composition and the caloric density were controlled, changes in de novo lipogenesis under these dietary conditions represent adaptation to changes in the fat-to-carbohydrate ratio of the diet. De novo lipogenesis was normally suppressed in response to the high-fat diet in the ZL rat to maintain a relatively constant amount of lipids transported. The ZDF rat had a higher rate of lipogenesis, which was not suppressed by the high-fat diet. The results suggest an important hormonal role of leptin in the feedback regulation of lipogenesis.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Carbohidratos de la Dieta/farmacología , Grasas de la Dieta/farmacología , Leptina/metabolismo , Lípidos/biosíntesis , Animales , Glucemia/metabolismo , Peso Corporal , Colesterol/sangre , Deuterio/análisis , Deuterio/metabolismo , Diabetes Mellitus Experimental/sangre , Ingestión de Energía , Insulina/sangre , Masculino , Ácido Oléico/sangre , Palmitatos/sangre , Ratas , Ratas Zucker , Estearatos/sangre , Triglicéridos/sangre
12.
Nutr Cancer ; 36(2): 150-4, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10890024

RESUMEN

Metabolic control analysis predicts that stimulators of transketolase enzyme synthesis such as thiamin (vitamin B-1) support a high rate of nucleic acid ribose synthesis necessary for tumor cell survival, chemotherapy resistance, and proliferation. Metabolic control analysis also predicts that transketolase inhibitor drugs will have the opposite effect on tumor cells. This may have important implications in the nutrition and future treatment of patients with cancer.


Asunto(s)
Neoplasias/patología , Ácidos Nucleicos/biosíntesis , Tiamina/efectos adversos , Transcetolasa/antagonistas & inhibidores , Transcetolasa/metabolismo , División Celular , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
13.
Cancer Res ; 60(5): 1183-5, 2000 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-10728670

RESUMEN

The invasive transformation of A-459 lung epithelial carcinoma cells has been linked to the autocrine regulation of malignant phenotypic changes by transforming growth factor beta (TGF-beta). Here we demonstrate, using stable 13C glucose isotopes, that the transformed phenotype is characterized by decreased CO2 production via direct glucose oxidation but increased nucleic acid ribose synthesis through the nonoxidative reactions of the pentose cycle. Increased nucleic acid synthesis through the nonoxidative pentose cycle imparts the metabolic adaptation of nontransformed cells to the invasive phenotype that potentially explains the fundamental metabolic disturbance in tumor cells: highly increased nucleic acid synthesis despite hypoxia and decreased glucose oxidation.


Asunto(s)
Adenocarcinoma/metabolismo , Glucosa/metabolismo , Neoplasias Pulmonares/metabolismo , Ribosa/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adenocarcinoma/patología , Transformación Celular Neoplásica , Humanos , Neoplasias Pulmonares/patología , Oxidación-Reducción , Vía de Pentosa Fosfato , Células Tumorales Cultivadas
14.
FEBS Lett ; 456(1): 113-8, 1999 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-10452541

RESUMEN

Transketolase (TK) reactions play a crucial role in tumor cell nucleic acid ribose synthesis utilizing glucose carbons, yet, current cancer treatments do not target this central pathway. Experimentally, a dramatic decrease in tumor cell proliferation after the administration of the TK inhibitor oxythiamine (OT) was observed in several in vitro and in vivo tumor models. Here, we demonstrate that pentose cycle (PC) inhibitors, OT and dehydroepiandrosterone (DHEA), efficiently regulate the cell cycle and tumor proliferation processes. Increasing doses of OT or DHEA were administered by daily intraperitoneal injections to Ehrlich's ascites tumor hosting mice for 4 days. The tumor cell number and their cycle phase distribution profile were determined by DNA flow histograms. Tumors showed a dose dependent increase in their G0-G1 cell populations after both OT and DHEA treatment and a simultaneous decrease in cells advancing to the S and G2-M cell cycle phases. This effect of PC inhibitors was significant, OT was more effective than DHEA, both drugs acted synergistically in combination and no signs of direct cell or host toxicity were observed. Direct inhibition of PC reactions causes a G1 cell cycle arrest similar to that of 2-deoxyglucose treatment. However, no interference with cell energy production and cell toxicity is observed. PC inhibitors, specifically ones targeting TK, introduce a new target site for the development of future cancer therapies to inhibit glucose utilizing pathways selectively for nucleic acid production.


Asunto(s)
Carcinoma de Ehrlich/patología , Deshidroepiandrosterona/farmacología , Fase G1/efectos de los fármacos , Oxitiamina/farmacología , Pentosas/metabolismo , Animales , Antimetabolitos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Ehrlich/tratamiento farmacológico , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Deshidroepiandrosterona/toxicidad , Relación Dosis-Respuesta a Droga , Corazón/efectos de los fármacos , Riñón/efectos de los fármacos , Riñón/patología , Hígado/efectos de los fármacos , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Oxitiamina/toxicidad , Transcetolasa/efectos de los fármacos , Transcetolasa/metabolismo
15.
J Surg Res ; 79(2): 154-7, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9758731

RESUMEN

BACKGROUND: Dehydroepiandrosterone (DHEA), an androgen precursor, inhibits the induction of pancreatic cancer in some animal models. Our laboratory has previously demonstrated that the sulfated form of DHEA (DHAS), when administered by intraperitoneal injection, inhibits the growth of pancreatic cancer xenografts in nude mice. In the present study, we hypothesize that DHEA-mediated pancreatic cancer growth inhibition is associated with alterations in plasma sex hormone concentrations. MATERIALS AND METHODS: Forty male, nude, athymic mice were fed either Teklad 22/5 rodent diet or diet supplemented with 0.6% DHEA ad libitum. Four weeks following the institution of the experimental diets, 1 x 10(6) MiaPaCa-2 cells were injected into the right flank of each animal. Tumor area was recorded weekly and tumor weights were measured after 5 weeks. Plasma DHAS, testosterone, and progesterone concentrations were determined by radioimmunoassay. RESULTS: Plasma DHAS, testosterone, and progesterone concentrations were all significantly elevated in the DHEA-treated group. DHEA-treated mouse plasma DHAS concentrations were approximately 50-fold higher than controls. Mean tumor weight was significantly reduced in the DHEA group (68.9 +/- 39.1 vs 121.0 +/- 64.3). DHEA treatment did not result in significant animal weight reductions and toxic side effects were not observed. CONCLUSIONS: Dietary supplementation with 0.6% DHEA causes significant elevations in plasma DHAS concentration. DHEA administration significantly inhibits pancreatic cancer cell growth at plasma concentrations 1 x 10(5)-fold lower than previously reported. The mechanism of action may involve elevated concentrations of sex hormones.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Deshidroepiandrosterona/administración & dosificación , Neoplasias Pancreáticas/patología , Adyuvantes Inmunológicos/farmacología , Administración Oral , Animales , Peso Corporal/efectos de los fármacos , División Celular/efectos de los fármacos , Deshidroepiandrosterona/farmacología , Sulfato de Deshidroepiandrosterona/sangre , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Pancreáticas/sangre , Progesterona/sangre , Valores de Referencia , Testosterona/sangre
16.
Med Hypotheses ; 50(6): 501-6, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9710324

RESUMEN

Long-acting somatostatin analogs have recently become supplemental drugs in the treatment of neurofibroma because of their marked tumor growth inhibitory effect. Somatostatin is currently under extended evaluation in other cancers as a possible supplemental drug to the treatment protocols in use. The mode of action is not known. Somatostatin has been shown to cause glucose intolerance by inhibiting glucose-6-phosphate dehydrogenase (G6PD) in fish liver. Recent data generated in our laboratory indicate that it is this pathway and the transketolase reactions of the pentose cycle (PC) which are directly involved in the ribose synthesis process of pancreatic adenocarcinoma cells. In cell culture, somatostatin alone inhibited glucose carbon recycling through the PC by 5.7%, which was increased to 19.8% in combination with oxythiamine, a competitive inhibitor of transketolase. Oxythiamine produced strong apoptosis in in-vitro hosted tumor cells. We hypothesize that somatostatin- and oxythiamine-induced antiproliferative action is mediated by the inhibition of G6PD, transketolase, or both.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Vía de Pentosa Fosfato/efectos de los fármacos , Somatostatina/farmacología , Animales , División Celular/efectos de los fármacos , Glucosa/metabolismo , Glucosafosfato Deshidrogenasa/metabolismo , Humanos , Modelos Biológicos , Transcetolasa/metabolismo
18.
Am J Physiol ; 274(5): E843-51, 1998 05.
Artículo en Inglés | MEDLINE | ID: mdl-9612242

RESUMEN

We present a single-tracer method for the study of the pentose phosphate pathway (PPP) using [1,2-13C2]glucose and mass isotopomer analysis. The metabolism of [1,2-13C2]glucose by the glucose-6-phosphate dehydrogenase, transketolase (TK), and transaldolase (TA) reactions results in unique pentose and lactate isotopomers with either one or two 13C substitutions. The distribution of these isotopomers was used to estimate parameters of the PPP using the model of Katz and Rognstad (J. Katz and R. Rognstad. Biochemistry 6: 2227-2247, 1967). Mass and position isotopomers of ribose, and lactate and palmitate (products from triose phosphate) from human hepatoma cells (Hep G2) incubated with 30% enriched [1,2-13C2]glucose were determined using gas chromatography-mass spectrometry. After 24-72 h incubation, 1.9% of lactate molecules in the medium contained one 13C substitution (m1) and 10% contained two 13C substitutions (m2). A similar m1-to-m2 ratio was found in palmitate as expected. Pentose cycle (PC) activity determined from incubation with [1,2-13C2]glucose was 5.73 +/- 0.52% of the glucose flux, which was identical to the value of PC (5.55 +/- 0.73%) determined by separate incubations with [1-13C] and [6-13C]glucose, 13C was found to be distributed in four ribose isotopomers ([1-13C]-, [5-13C]-, [1,2-13C2]-, and [4,5-13C2]ribose). The observed ribose isotopomer distribution was best matched with that provided from simulation by substituting 0.032 for TK and 0.85 for TA activity relative to glucose uptake into the model of Katz and Rognstad. The use of [1,2-13C2]glucose not only permits the determination of PC but also allows estimation of relative rates through the TK and TA reactions.


Asunto(s)
Glucosa/metabolismo , Vía de Pentosa Fosfato/fisiología , Isótopos de Carbono , Glucosafosfato Deshidrogenasa/metabolismo , Humanos , Ácido Láctico/metabolismo , Palmitatos/metabolismo , Pentosas/metabolismo , Ribosa/metabolismo , Transaldolasa/metabolismo , Transcetolasa/metabolismo , Células Tumorales Cultivadas
19.
Anticancer Res ; 18(1B): 595-602, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9568183

RESUMEN

The objectives of this review are to (a) explain the mechanism by which thiamine (vitamin B1) promotes nucleic acid ribose synthesis and tumor cell proliferation via the nonoxidative transketolase (TK) pathway; (b) estimate the thiamine intake of cancer patients and (c) provide background information and to develop guidelines for alternative treatments with antithiamine transketolase inhibitors in the clinical setting. Clinical and experimental data demonstrate increased thiamine utilization of human tumors and its interference with experimental chemotherapy. Analysis of RNA ribose indicates that glucose carbons contribute to over 90% of ribose synthesis in cultured cervix und pancreatic carcinoma cells and that ribose is synthesized primarily through the thiamine dependent TK pathway (> 70%). Antithiamine compounds significantly inhibit nucleic acid synthesis and tumor cell proliferation in vitro and in vivo in several tumor models. The medical literature reveals little information regarding the role of the thiamine dependent TK reaction in tumor cell ribose production which is a central process in de novo nucleic acid synthesis and the salvage pathways for purines. Consequently, current thiamine administration protocols oversupply thiamine by 200% to 20,000% of the recommended dietary allowance, because it is considered harmless and needed by cancer patients. The thiamine dependent TK pathway is the central avenue which supplies ribose phosphate for nucleic acids in tumors and excessive thiamine supplementation maybe responsible for failed therapeutic attempts to terminate cancer cell proliferation. Limited administration of thiamine and concomitant treatment with transketolase inhibitors is a more rational approach to treat cancer.


Asunto(s)
Neoplasias/tratamiento farmacológico , Tiamina/efectos adversos , Tiamina/uso terapéutico , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , División Celular , Guías como Asunto , Humanos , Ácidos Nucleicos/biosíntesis , Tiamina/química , Transcetolasa/metabolismo
20.
Surgery ; 123(3): 315-20, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9526524

RESUMEN

BACKGROUND: Streptozotocin-diabetes prevents induction of pancreatic tumors in several animal models and inhibits the growth of established human pancreatic cancer implants in nude mice. However, it also promotes growth of the hamster pancreatic cancer cell line, H2T, in the Syrian hamster. To test the hypothesis that these contradictory effects are due to tumor host differences, the growth of the H2T cell line was examined in the streptozotocin-diabetic nude mouse. METHODS: H2T cells were implanted subcutaneously into streptozotocin-diabetic nude mice (n = 10) and untreated control mice (n = 10). After 21 days, tumors were excised and weighed. Plasma insulin and somatostatin were determined by radioimmunoassay. RESULTS: After 3 weeks, tumors in the control group weighed 118 mg and tumors in the diabetic group weighed 28 mg (p < 0.001). Plasma insulin was significantly decreased in the streptozotocin-treated animals compared with control animals (insulin, 23 microU/ml vs 31 microU/ml; p < 0.001). In contrast, somatostatin was significantly elevated in the streptozotocin-diabetic group compared with the control group (somatostatin, 179 pg/ml versus 54 pg/ml, p < 0.001). Competitive binding studies revealed specific cell surface receptors for insulin (Kd, 15.5 nmol/L), and somatostatin (Kd, 2.5 nmol/L) on the H2T cells. In an in vitro cell proliferation assay, cell division was promoted by insulin (p < 0.01, maximum +11%) and inhibited by somatostatin (p < 0.01, maximum -18%). CONCLUSIONS: The variable effect of streptozotocin-diabetes on pancreatic cancer growth is due to differences in the tumor host. The growth of pancreatic cancer, particularly in streptozotocin-diabetic nude mice, may be influenced by gut peptides in a receptor-dependent fashion.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Neoplasias Pancreáticas/patología , Animales , Peso Corporal , Cricetinae , Mesocricetus , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Pancreáticas/complicaciones , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Receptores de Glucagón/metabolismo , Receptores de Somatostatina/metabolismo , Trasplante Heterólogo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA