15 research outputs found

    Age at symptom onset and death and disease duration in genetic frontotemporal dementia : an international retrospective cohort study

    Get PDF
    Background: Frontotemporal dementia is a heterogenous neurodegenerative disorder, with about a third of cases being genetic. Most of this genetic component is accounted for by mutations in GRN, MAPT, and C9orf72. In this study, we aimed to complement previous phenotypic studies by doing an international study of age at symptom onset, age at death, and disease duration in individuals with mutations in GRN, MAPT, and C9orf72. Methods: In this international, retrospective cohort study, we collected data on age at symptom onset, age at death, and disease duration for patients with pathogenic mutations in the GRN and MAPT genes and pathological expansions in the C9orf72 gene through the Frontotemporal Dementia Prevention Initiative and from published papers. We used mixed effects models to explore differences in age at onset, age at death, and disease duration between genetic groups and individual mutations. We also assessed correlations between the age at onset and at death of each individual and the age at onset and at death of their parents and the mean age at onset and at death of their family members. Lastly, we used mixed effects models to investigate the extent to which variability in age at onset and at death could be accounted for by family membership and the specific mutation carried. Findings: Data were available from 3403 individuals from 1492 families: 1433 with C9orf72 expansions (755 families), 1179 with GRN mutations (483 families, 130 different mutations), and 791 with MAPT mutations (254 families, 67 different mutations). Mean age at symptom onset and at death was 49\ub75 years (SD 10\ub70; onset) and 58\ub75 years (11\ub73; death) in the MAPT group, 58\ub72 years (9\ub78; onset) and 65\ub73 years (10\ub79; death) in the C9orf72 group, and 61\ub73 years (8\ub78; onset) and 68\ub78 years (9\ub77; death) in the GRN group. Mean disease duration was 6\ub74 years (SD 4\ub79) in the C9orf72 group, 7\ub71 years (3\ub79) in the GRN group, and 9\ub73 years (6\ub74) in the MAPT group. Individual age at onset and at death was significantly correlated with both parental age at onset and at death and with mean family age at onset and at death in all three groups, with a stronger correlation observed in the MAPT group (r=0\ub745 between individual and parental age at onset, r=0\ub763 between individual and mean family age at onset, r=0\ub758 between individual and parental age at death, and r=0\ub769 between individual and mean family age at death) than in either the C9orf72 group (r=0\ub732 individual and parental age at onset, r=0\ub736 individual and mean family age at onset, r=0\ub738 individual and parental age at death, and r=0\ub740 individual and mean family age at death) or the GRN group (r=0\ub722 individual and parental age at onset, r=0\ub718 individual and mean family age at onset, r=0\ub722 individual and parental age at death, and r=0\ub732 individual and mean family age at death). Modelling showed that the variability in age at onset and at death in the MAPT group was explained partly by the specific mutation (48%, 95% CI 35\u201362, for age at onset; 61%, 47\u201373, for age at death), and even more by family membership (66%, 56\u201375, for age at onset; 74%, 65\u201382, for age at death). In the GRN group, only 2% (0\u201310) of the variability of age at onset and 9% (3\u201321) of that of age of death was explained by the specific mutation, whereas 14% (9\u201322) of the variability of age at onset and 20% (12\u201330) of that of age at death was explained by family membership. In the C9orf72 group, family membership explained 17% (11\u201326) of the variability of age at onset and 19% (12\u201329) of that of age at death. Interpretation: Our study showed that age at symptom onset and at death of people with genetic frontotemporal dementia is influenced by genetic group and, particularly for MAPT mutations, by the specific mutation carried and by family membership. Although estimation of age at onset will be an important factor in future pre-symptomatic therapeutic trials for all three genetic groups, our study suggests that data from other members of the family will be particularly helpful only for individuals with MAPT mutations. Further work in identifying both genetic and environmental factors that modify phenotype in all groups will be important to improve such estimates. Funding: UK Medical Research Council, National Institute for Health Research, and Alzheimer's Society

    Simvastatin Inducing Pc3 Prostate Cancer Cell Necrosis Mediated By Calcineurin And Mitochondrial Dysfunction

    No full text
    In the present study we analyzed the mechanisms of simvastatin toxicity for the PC3 human prostate cancer cell line. At 10 μM, simvastatin induced principally apoptosis, which was prevented by mevalonic acid but not by cyclosporin A, the inhibitor of calcineurin and mitochondrial permeability transition (MPT). At 60 μM, simvastatin induced the necrosis of PC3 cells insensitive to mevalonic acid. Cell necrosis was preceded by a threefold increase in cytosolic free Ca 2+ concentration and a significant decrease in both respiration rate and mitochondrial membrane potential. Both mitochondrial dysfunction and necrosis were sensitive to the compounds cyclosporin A and bongkrekic acid, as well as the calcineurin inhibitor FK506. We have concluded that simvastatin-induced PC3 cells apoptosis is dependent on 3-hydroxy-3-methylglutaryl coenzyme-A reductase inhibition and independent of MPT, whereas necrosis is dependent on mitochondrial dysfunction caused, at least in part, by calcineurin. © 2008 Springer Science+Business Media, LLC.404307314Almeida, S., Domingues, A., Rodrigues, L., (2004) Neurobiol Dis, 17, pp. 435-444Ankarcrona, M., Dypbukt, J.M., Orrenius, S., (1996) FEBS Lett, 394, pp. 321-324Campos, C.B.L., Degasperi, G.R., Pacífico, D.S., (2004) Biochem Pharmacol, 68, pp. 2197-2206Collins, R., Armitage, J., Parish, S., (2003) Lancet, 361, pp. 2005-2016Dawson, T.M., Steiner, J.P., Dawson, V.L., (1993) Proc Natl Acad Sci U S a, 90, pp. 9808-9812Degasperi, G.R., Velho, J.A., Zecchin, K.G., (2006) J Bioenerg Biomembr, 38, pp. 1-10Demierre, M.-F., Higgins, P.D.R., Gruber, S.B., (2005) Nat Rev Cancer, 5, pp. 930-942Ghosh, P.M., Ghosh-Choudhury, N., Moyer, M.L., (1999) Oncogene, 18, pp. 4120-4130Gunter, K.K., Gunter, T.E., (1994) J Bioenerg Biomembr, 26, pp. 471-485Halestrap, A.P., Brenner, C., (2003) Curr Med Chem, 10, pp. 1507-1525Halestrap, A.P., Connern, C.P., Griffths, E.J., (1997) Mol Cell Biochem, 174, pp. 167-172Hara, M.R., Snyder, S.H., (2007) Toxicol, 47, pp. 117-141Hindler, K., Cleeland, C.S., Rivera, E., (2006) Oncologist, 11, pp. 306-315Holden, M.J., Sze, H., (1989) Plant Physiol, 91, pp. 1296-1302Hoque, A., Chen, H., Xu, X.C., (2008) Cancer Epidemiol Biomarkers Prev, 17, pp. 88-94Kowaltowski, A.J., Castilho, R.F., Vercesi, A.E., (2001) FEBS Lett, 495, pp. 12-15Kroemer, G., Galluzi, L., Brenner, C., (2007) Physiol Rev, 87, pp. 99-163Li, Y.C., Park, M.J., Ye, S.-K., (2006) Am J Pathol, 168, pp. 1107-1118Liu, J., Farmer, J.D.J., Lane, W.S., Friedman, J., (1991) Cell, 66, pp. 807-815Manev, H., Favaron, M., Candeo, P., (1993) Brain Res, 624, pp. 331-335Marcelli, M., Glenn, R.C., Haidacher, S.J., (1998) Cancer Res, 58, pp. 76-83Park, C., Lee, I., Kang, W.K., (2001) Carcinogenesis, 22, pp. 1727-1731Platz, E.A., Leitzmann, M.F., Visvanathan, K., (2006) J Natl Cancer Inst, 98, pp. 1819-1825Rao, S., Porter, D.C., Chen, X., (1999) Proc Natl Acad Sci U S a, 96, pp. 7797-7802Shepherd, J., Cobbe, S.M., Ford, I., (1995) N Engl J Med, 333, pp. 1301-1307Shibasaki, F., McKeon, F., (1995) Cell Biol, 131, pp. 735-743Sivaprasad, U., Abbas, T., Dutta, A., (2006) Mol Cancer Ther, 5, pp. 2310-2316Springer, J.E., Azbill, R.D., Nottingham, S.A., (2000) J Neurosci, 20, pp. 7246-7251Ukomadu, C., Dutta, A., (2003) J Biol Chem, 278, pp. 4840-4846Velho, J.A., Okanobo, H., Degasperi, G.R., (2006) Toxicology, 219, pp. 124-132Vercesi, A.E., Kowaltowski, A.J., Oliveira, H.C.F., (2006) Front Biosc, 11, pp. 2554-2564Wang, H.G., Pathan, N., Ethell, I.M., (1999) Science, 284, pp. 339-343Weitz-Schmidt, G., Welzenbach, K., Brinkmann, V., (2001) Nat Med, 7, pp. 687-692Zecchin, K.G., Seidinger, A.L.O., Degasperi, G.R., (2007) J Bioenerg Biomembr, 39, pp. 186-19

    Protection Of Rat Skeletal Muscle Fibers By Either L-carnitine Or Coenzyme Q10 Against Statins Toxicity Mediated By Mitochondrial Reactive Oxygen Generation

    No full text
    Mitochondrial redox imbalance has been implicated in mechanisms of aging, various degenerative diseases and drug-induced toxicity. Statins are safe and well-tolerated therapeutic drugs that occasionally induce myotoxicity such as myopathy and rhabdomyolysis. Previous studies indicate that myotoxicity caused by statins may be linked to impairment of mitochondrial functions. Here, we report that 1-h incubation of permeabilized rat soleus muscle fiber biopsies with increasing concentrations of simvastatin (1-40 μM) slowed the rates of ADP-or FCCP-stimulated respiration supported by glutamate/malate in a dose-dependent manner, but caused no changes in resting respiration rates. Simvastatin (1 μM) also inhibited the ADP-stimulated mitochondrial respiration supported by succinate by 24% but not by TMPD/ascorbate. Compatible with inhibition of respiration, 1 μM simvastatin stimulated lactate release from soleus muscle samples by 26%. Co-incubation of muscle samples with 1 mM L-carnitine, 100 μM mevalonate or 10 μM coenzyme Q10 (Co-Q10) abolished simvastatin effects on both mitochondrial glutamate/malate-supported respiration and lactate release. Simvastatin (1 μM) also caused a 2-fold increase in the rate of hydrogen peroxide generation and a decrease in Co-Q10 content by 44%. Mevalonate, Co-Q10 or L-carnitine protected against stimulation of hydrogen peroxide generation but only mevalonate prevented the decrease in Co-Q10 content. Thus, independently of Co-Q10 levels, L-carnitine prevented the toxic effects of simvastatin. This suggests that mitochondrial respiratory dysfunction induced by simvastatin, is associated with increased generation of superoxide, at the levels of complexes-I and II of the respiratory chain. In all cases the damage to these complexes, presumably at the level of 4Fe-4S clusters, is prevented by L-carnitine. © 2013 La Guardia, Alberici, Ravagnani, Catharino and Vercesi.4 MAYAnderson, E.J., Neufer, P.D., Type II skeletal myofibers possess unique properties that potentiate mitochondrial H(2)O(2) generation (2006) Am. J. Physiol. Cell Physiol., 290, pp. C844-C851Arduini, A., Peschechera, A., Giannessi, F., Carminati, P., Improvement of statin-associated myotoxicity by L-carnitine (2004) J. Thromb. Haemost., 2, pp. 2270-2271Benati, D., Ferro, M., Savino, M.T., Ulivieri, C., Schiavo, E., Nuccitelli, A., Opposite effects of simvastatin on the bactericidal and inflammatory response of macrophages to opsonized S. aureus (2010) J. Leukoc. Biol., 87, pp. 433-442Binienda, Z.K., Neuroprotective effects of L-carnitine in induced mitochondrial dysfunction (2003) Ann. N. Y. Acad. Sci., 993, pp. 289-295. , discussion: 345-349Binienda, Z., Przybyla-Zawislak, B., Virmani, A., Schmued, L., L-carnitine and neuroprotection in the animal model of mitochondrial dysfunction (2005) Ann. N. Y. Acad. Sci., 1053, pp. 174-182Bookstaver, D.A., Burkhalter, N.A., Hatzigeorgiou, C., Effect of coenzyme Q10 supplementation on statin-induced myalgias (2012) Am. J. Cardiol., 110, pp. 526-529Bouitbir, J., Charles, A.L., Echaniz-Laguna, A., Kindo, M., Daussin, F., Auwerx, J., Opposite effects of statins on mitochondria of cardiac and skeletal muscles: a 'mitohormesis' mechanism involving reactive oxygen species and PGC-1 (2012) Eur. Heart J., 33, pp. 1397-1407Bouitbir, J., Daussin, F., Charles, A.L., Rasseneur, L., Dufour, S., Richard, R., Mitochondria of trained skeletal muscle are protected from deleterious effects of statins (2012) Muscle Nerve, 46, pp. 367-373Bouton, C., Raveau, M., Drapier, J.C., Modulation of iron regulatory protein functions (1996) Further insights into the role of nitrogen- and oxygen-derived reactive species. J. Biol. Chem., 271, pp. 2300-2306Bruckert, E., Hayem, G., Dejager, S., Yau, C., Begaud, B., Mild to moderate muscular symptoms with high-dosage statin therapy in hyperlipidemic patients-the PRIMO study (2005) Cardiovasc. Drugs Ther., 19, pp. 403-414Castilho, R.F., Kowaltowski, A.J., Meinicke, A.R., Vercesi, A.E., Oxidative damage of mitochondria induced by Fe(II)citrate or t-butyl hydroperoxide in the presence of Ca2+: effect of coenzyme Q redox state (1995) Free Radic. Biol. Med., 18, pp. 55-59Chariot, P., Monnet, I., Mouchet, M., Rohr, M., Lefaucheur, J.P., Dubreuil-Lemaire, M.L., Determination of the blood lactate:pyruvate ratio as a noninvasive test for the diagnosis of zidovudine myopathy (1994) Arthritis Rheum, 37, pp. 583-586Costa, R.A.P., Fernandes, M.P., Souza-Pinto, N.C.D., Vercesi, A.E., Protective effects of L-carnitine and piracetam against mitochondrial permeability transition and PC3 cell necrosis induced by simvastatin (2013) Eur. J. Pharmacol., 701, pp. 82-86Deichmann, R., Lavie, C., Andrews, S., Coenzyme q10 and statin-induced mitochondrial dysfunction (2010) Ochsner J, 10, pp. 16-21Demicheli, V., Quijano, C., Alvarez, B., Radi, R., Inactivation and nitration of human superoxide dismutase (SOD) by fluxes of nitric oxide and superoxide (2007) Free Radic. Biol. Med., 42, pp. 1359-1368De Pinieux, G., Chariot, P., Ammi-Said, M., Louarn, F., Lejonc, J.L., Astier, A., Lipid-lowering drugs and mitochondrial function: effects of HMG-CoA reductase inhibitors on serum ubiquinone and blood lactate/pyruvate ratio (1996) Br. J. Clin. Pharmacol., 42, pp. 333-337Elinos-Calderon, D., Robledo-Arratia, Y., Perez-De La Cruz, V., Pedraza-Chaverri, J., Ali, S.F., Santamaria, A., Early nerve ending rescue from oxidative damage and energy failure by L: - carnitine as post-treatment in two neurotoxic models in rat: recovery of antioxidant and reductive capacities. Exp (2009) Brain Res, 197, pp. 287-296Endo, A., The discovery and development of HMG-CoA reductase inhibitors (1992) J. Lipid Res., 33, pp. 1569-1582Figueira, T.R., Barros, M.H., Camargo, A.A., Castilho, R.F., Ferreira, J.C., Kowaltowski, A.J., Mitochondria as a source of reactive oxygen and nitrogen species: from molecular mechanisms to human health (2013) Antioxid. Redox Signal., 18, pp. 2029-2074Flint, D.H., Tuminello, J.F., Emptage, M.H., The inactivation of Fe-S cluster containing hydro-lyases by superoxide (1993) J. Biol. Chem., 268, pp. 22369-22376Fridovich, I., Superoxide radical and superoxide dismutases (1995) Annu. Rev. Biochem., 64, pp. 97-112Ghirlanda, G., Oradei, A., Manto, A., Lippa, S., Uccioli, L., Caputo, S., Evidence of plasma CoQ10-lowering effect by HMG-CoA reductase inhibitors: a double-blind, placebo-controlled study (1993) J. Clin. Pharmacol., 33, pp. 226-229Graham, D., Huynh, N.N., Hamilton, C.A., Beattie, E., Smith, R.A., Cocheme, H.M., Mitochondria-targeted antioxidant MitoQ10 improves endothelial function and attenuates cardiac hypertrophy (2009) Hypertension, 54, pp. 322-328Grijalba, M.T., Vercesi, A.E., Schreier, S., Ca2+-induced increased lipid packing and domain formation in submitochondrial particles (1999) A possible early step in the mechanism of Ca2+-stimulated generation of reactive oxygen species by the respiratory chain. Biochemistry, 38, pp. 13279-13287Gulcin, I., Antioxidant and antiradical activities of L-carnitine (2006) Life Sci, 78, pp. 803-811Itagaki, M., Takaguri, A., Kano, S., Kaneta, S., Ichihara, K., Satoh, K., Possible mechanisms underlying statin-induced skeletal muscle toxicity in L6 fibroblasts and in rats (2009) J. Pharmacol. Sci., 109, pp. 94-101Kaikkonen, J., Tuomainen, T.P., Nyyssonen, K., Salonen, J.T., Coenzyme Q10 absorption, antioxidative properties, determinants, and plasma levels (2002) Free Radic. Res, 36, pp. 389-397Kaufmann, P., Torok, M., Zahno, A., Waldhauser, K.M., Brecht, K., Krahenbuhl, S., Toxicity of statins on rat skeletal muscle mitochondria (2006) Cell. Mol. Life Sci., 63, pp. 2415-2425Keil, U., Scherping, I., Hauptmann, S., Schuessel, K., Eckert, A., Muller, W.E., Piracetam improves mitochondrial dysfunction following oxidative stress (2006) Br. J. Pharmacol., 147, pp. 199-208Kettawan, A., Takahashi, T., Kongkachuichai, R., Charoenkiatkul, S., Kishi, T., Okamoto, T., Protective effects of coenzyme q(10) on decreased oxidative stress resistance induced by simvastatin (2007) J. Clin. Biochem. Nutr., 40, pp. 194-202Kumaran, S., Savitha, S., Anusuya Devi, M., Panneerselvam, C., L-carnitine and DL-alpha-lipoic acid reverse the age-related deficit in glutathione redox state in skeletal muscle and heart tissues (2004) Mech. Ageing Dev., 125, pp. 507-512Kumaran, S., Subathra, M., Balu, M., Panneerselvam, C., Supplementation of L-carnitine improves mitochondrial enzymes in heart and skeletal muscle of aged rats (2005) Exp. Aging Res., 31, pp. 55-67Kuznetsov, A.V., Veksler, V., Gellerich, F.N., Saks, V., Margreiter, R., Kunz, W.S., Analysis of mitochondrial function in situ in permeabilized muscle fibers, tissues and cells (2008) Nat. Protoc., 3, pp. 965-976Kwak, H.B., Thalacker-Mercer, A., Anderson, E.J., Lin, C.T., Kane, D.A., Lee, N.S., Simvastatin impairs ADP-stimulated respiration and increases mitochondrial oxidative stress in primary human skeletal myotubes (2012) Free Radic. Biol. Med., 52, pp. 198-207Laaksonen, R., Jokelainen, K., Sahi, T., Tikkanen, M.J., Himberg, J.J., Decreases in serum ubiquinone concentrations do not result in reduced levels in muscle tissue during short-term simvastatin treatment in humans (1995) Clin. Pharmacol. Ther., 57, pp. 62-66Prevention of cardiovascular events and death with pravastatin in patients with coronary heart disease and a broad range of initial cholesterol levels (1998) N. Engl. J. Med., 339, pp. 1349-1357. , LIPID (The Long-Term Intervention with Pravastatin in Ischaemic Disease) Study GroupLittarru, G.P., Langsjoen, P., Coenzyme Q10 and statins: biochemical and clinical implications (2007) Mitochondrion, 7 (SUPPL.), pp. S168-S174Littarru, G.P., Tiano, L., Clinical aspects of coenzyme Q10 an update (2010) Nutrition, 26, pp. 250-254Mabuchi, H., Nohara, A., Kobayashi, J., Kawashiri, M.A., Katsuda, S., Inazu, A., Effects of CoQ10 supplementation on plasma lipoprotein lipid (2007) CoQ10 and liver and muscle enzyme levels in hypercholesterolemic patients treated with atorvastatin: a randomized double-blind study. Atherosclerosis, 195, pp. e182-e189Mescka, C., Moraes, T., Rosa, A., Mazzola, P., Piccoli, B., Jacques, C., In vivo neuroprotective effect of L-carnitine against oxidative stress in maple syrup urine disease (2011) Metab. Brain Dis., 26, pp. 21-28Miyake, Y., Shouzu, A., Nishikawa, M., Yonemoto, T., Shimizu, H., Omoto, S., Effect of treatment with 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors on serum coenzyme Q10 in diabetic patients (1999) Arzneimittelforschung, 49, pp. 324-329Moretti, S., Famularo, G., Marcellini, S., Boschini, A., Santini, G., Trinchieri, V., L-carnitine reduces lymphocyte apoptosis and oxidant stress in HIV-1-infected subjects treated with zidovudine and didanosine (2002) Antioxid. Redox Signal., 4, pp. 391-403Munnich, A., Rustin, P., Rotig, A., Chretien, D., Bonnefont, J.P., Nuttin, C., Clinical aspects of mitochondrial disorders (1992) J. Inherit. Metab. Dis., 15, pp. 448-455Nadanaciva, S., Dykens, J.A., Bernal, A., Capaldi, R.A., Will, Y., Mitochondrial impairment by PPAR agonists and statins identified via immunocaptured OXPHOS complex activities and respiration (2007) Toxicol. Appl. Pharmacol., 223, pp. 277-287Naini, A., Lewis, V.J., Hirano, M., Dimauro, S., Primary coenzyme Q10 deficiency and the brain (2003) Biofactors, 18, pp. 145-152Oliveira, K.A., Zecchin, K.G., Alberici, L.C., Castilho, R.F., Vercesi, A.E., Simvastatin inducing PC3 prostate cancer cell necrosis mediated by calcineurin and mitochondrial dysfunction (2008) J. Bioenerg. Biomembr., 40, pp. 307-314Paiva, H., Thelen, K.M., Van Coster, R., Smet, J., De Paepe, B., Mattila, K.M., High-dose statins and skeletal muscle metabolism in humans: a randomized, controlled trial (2005) Clin. Pharmacol. Ther., 78, pp. 60-68Panov, A., Dikalov, S., Shalbuyeva, N., Taylor, G., Sherer, T., Greenamyre, J.T., Rotenone model of Parkinson disease: multiple brain mitochondria dysfunctions after short term systemic rotenone intoxication (2005) J. Biol. Chem., 280, pp. 42026-42035Radi, R., Rodriguez, M., Castro, L., Telleri, R., Inhibition of mitochondrial electron transport by peroxynitrite (1994) Arch. Biochem. Biophys., 308, pp. 89-95Redfearn, E.R., Whittaker, P.A., The determination of the oxidation-reduction states of ubiquinone (coenzyme Q) in rat-liver mitochondria (1966) Biochim. Biophys. Acta, 118, pp. 413-418Robinson, B.H., Lacticacidemia. Biochemical, clinical, and genetic considerations (1989) Adv. Hum. Genet, 18, pp. 151-179. , 371-372Rundek, T., Naini, A., Sacco, R., Coates, K., Dimauro, S., Atorvastatin decreases the coenzyme Q10 level in the blood of patients at risk for cardiovascular disease and stroke (2004) Arch. Neurol., 61, pp. 889-892Sacher, J., Weigl, L., Werner, M., Szegedi, C., Hohenegger, M., Delineation of myotoxicity induced by 3-hydroxy-3-methylglutaryl CoA reductase inhibitors in human skeletal muscle cells (2005) J. Pharmacol. Exp. Ther., 314, pp. 1032-1041Sener, G., Paskaloglu, K., Satiroglu, H., Alican, I., Kacmaz, A., Sakarcan, A., L-carnitine ameliorates oxidative damage due to chronic renal failure in rats (2004) J. Cardiovasc. Pharmacol., 43, pp. 698-705Shen, W., Liu, K., Tian, C., Yang, L., Li, X., Ren, J., Protective effects of R-alpha-lipoic acid and acetyl-L-carnitine in MIN6 and isolated rat islet cells chronically exposed to oleic acid (2008) J. Cell. Biochem., 104, pp. 1232-1243Shepherd, D., Garland, P.B., The kinetic properties of citrate synthase from rat liver mitochondria (1969) Biochem. J., 114, pp. 597-610Silva-Adaya, D., Perez-De La Cruz, V., Herrera-Mundo, M.N., Mendoza-Macedo, K., Villeda-Hernandez, J., Binienda, Z., Excitotoxic damage, disrupted energy metabolism, and oxidative stress in the rat brain: antioxidant and neuroprotective effects of L-carnitine (2008) J. Neurochem., 105, pp. 677-689Sirvent, P., Bordenave, S., Vermaelen, M., Roels, B., Vassort, G., Mercier, J., Simvastatin induces impairment in skeletal muscle while heart is protected (2005) Biochem. Biophys. Res. Commun., 338, pp. 1426-1434Sirvent, P., Mercier, J., Vassort, G., Lacampagne, A., Simvastatin triggers mitochondria-induced Ca2+ signaling alteration in skeletal muscle (2005) Biochem. Biophys. Res. Commun., 329, pp. 1067-1075Sirvent, P., Fabre, O., Bordenave, S., Hillaire-Buys, D., Raynaud De Mauverger, E., Lacampagne, A., Muscle mitochondrial metabolism and calcium signaling impairment in patients treated with statins (2012) Toxicol. Appl. Pharmacol., 259, pp. 263-268Sirvent, P., Mercier, J., Lacampagne, A., New insights into mechanisms of statin-associated myotoxicity (2008) Curr. Opin. Pharmacol., 8, pp. 333-338Skottheim, I.B., Gedde-Dahl, A., Hejazifar, S., Hoel, K., Asberg, A., Statin induced myotoxicity: the lactone forms are more potent than the acid forms in human skeletal muscle cells in vitro (2008) Eur. J. Pharm. Sci., 33, pp. 317-325Thibault, A., Samid, D., Tompkins, A.C., Figg, W.D., Cooper, M.R., Hohl, R.J., Phase I study of lovastatin, an inhibitor of the mevalonate pathway, in patients with cancer (1996) Clin. Cancer Res, 2, pp. 483-491Tobert, J.A., Hitzenberger, G., Kukovetz, W.R., Holmes, I.B., Jones, K.H., Rapid and substantial lowering of human serum cholesterol by mevinolin (MK-803), an inhibitor of hydroxymethylglutaryl-coenzyme A reductase (1982) Atherosclerosis, 41, pp. 61-65Vamos, E., Voros, K., Vecsei, L., Klivenyi, P., Neuroprotective effects of L-carnitine in a transgenic animal model of Huntington's disease (2010) Biomed. Pharmacother., 64, pp. 282-286Velho, J.A., Okanobo, H., Degasperi, G.R., Matsumoto, M.Y., Alberici, L.C., Cosso, R.G., Statins induce calcium-dependent mitochondrial permeability transition (2006) Toxicology, 219, pp. 124-132Venero, C.V., Thompson, P.D., Managing statin myopathy (2009) Endocrinol. Metab. Clin. North Am., 38, pp. 121-136Virmani, A., Gaetani, F., Binienda, Z., Xu, A., Duhart, H., Ali, S.F., Role of mitochondrial dysfunction in neurotoxicity of MPP+: partial protection of PC12 cells by acetyl-L-carnitine (2004) Ann. N. Y. Acad. Sci., 1025, pp. 267-273Yapar, K., Kart, A., Karapehlivan, M., Atakisi, O., Tunca, R., Erginsoy, S., Hepatoprotective effect of L-carnitine against acute acetaminophen toxicity in mice (2007) Exp. Toxicol. Pathol., 59, pp. 121-128Yasuda, N., Matzno, S., Iwano, C., Nishikata, M., Matsuyama, K., Evaluation of apoptosis and necrosis induced by statins using fluorescence-enhanced flow cytometry (2005) J. Pharm. Biomed. Anal., 39, pp. 712-717Ye, J., Li, J., Yu, Y., Wei, Q., Deng, W., Yu, L., L-carnitine attenuates oxidant injury in HK-2 cells via ROS-mitochondria pathway (2010) Regul. Pept., 161, pp. 58-66Young, J.M., Molyneux, S.L., Reinheimer, A.M., Florkowski, C.M., Frampton, C.M., Scott, R.S., Relationship between plasma coenzyme Q10, asymmetric dimethylarginine and arterial stiffness in patients with phenotypic or genotypic familial hypercholesterolemia on long-term statin therapy (2011) Atherosclerosis, 218, pp. 188-193Zhang, Z., Zhao, M., Wang, J., Ding, Y., Dai, X., Li, Y., Effect of acetyl-L-carnitine on the insulin resistance of L6 cells induced by tumor necrosis factor-alpha (2010) Wei Sheng Yan Jiu, 39, pp. 152-15

    Administration Of A Murine Diet Supplemented With Conjugated Linoleic Acid Increases The Expression And Activity Of Hepatic Uncoupling Proteins

    No full text
    Daily intake of conjugated linoleic acid (CLA) has been shown to reduce body fat accumulation and to increase body metabolism; this latter effect has been often associated with the up-regulation of uncoupling proteins (UCPs). Here we addressed the effects of a CLA-supplemented murine diet (∼2 % CLA mixture, cis-9, trans-10 and trans-10, cis-12 isomers; 45 % of each isomer on alternating days) on mitochondrial energetics, UCP2 expression/activity in the liver and other associated morphological and functional parameters, in C57BL/6 mice. Diet supplementation with CLA reduced both lipid accumulation in adipose tissues and triacylglycerol plasma levels, but did not augment hepatic lipid storage. Livers of mice fed a diet supplemented with CLA showed high UCP2 mRNA levels and the isolated hepatic mitochondria showed indications of UCP activity: in the presence of guanosine diphosphate, the higher stimulation of respiration promoted by linoleic acid in mitochondria from the CLA mice was almost completely reduced to the level of the stimulation from the control mice. Despite the increased generation of reactive oxygen species through oxi-reduction reactions involving NAD+/NADH in the Krebs cycle, no oxidative stress was observed in the liver. In addition, in the absence of free fatty acids, basal respiration rates and the phosphorylating efficiency of mitochondria were preserved. These results indicate a beneficial and secure dose of CLA for diet supplementation in mice, which induces UCP2 overexpression and UCP activity in mitochondria while preserving the lipid composition and redox state of the liver. © Springer Science+Business Media, LLC 2012.445587596Alberici, L.C., Oliveira, H.C., Patrício, P.R., Kowaltowski, A.J., Vercesi, A.E., Hyperlipidemic mice present enhanced catabolism and higher mitochondrial ATP-sensitive K+channel activity (2006) Gastroenterology, 131, pp. 1228-1234Alberici, R.M., Simas, R.C., Sanvido, G.B., Romão, W., Lalli, P.M., Benassi, M., Ambient mass spectrometry: Bringing MS into the real world (2010) Anal Bioanal Chem, 398, pp. 265-294Alberici, L.C., Oliveira, H.C., Catharino, R.R., Vercesi, A.E., Eberlin, M.N., Alberici, R.M., Distinct hepatic lipid profile of hypertriglyceridemic mice determined by easy ambient sonic-spray ionization mass spectrometry (2011) Anal Bioanal Chem, 401, pp. 1651-1659Azain, M.J., Hausman, D.B., Sisk, M.B., Flatt, W.P., Jewell, D.E., Dietary conjugated linoleic acid reduces rat adipose tissue cell size rather than cell number (2000) J Nutr, 130, pp. 1548-1554Banni, S., Carta, G., Angioni, E., Murru, E., Scanu, P., Melis, M.P., Distribution of conjugated linoleic acid and metabolites in different lipid fractions in the rat liver (2001) J Lipid Res, 42, pp. 1056-1061Bligh, E.G., Dyer, W.J., A rapid method of total lipid extraction and purification (1959) Can J Biochem Physiol, 37, pp. 911-917Buege, J.A., Aust, S.D., Microsomal lipid peroxidation (1978) Methods Enzymol, 52, pp. 302-310Cherian, G., Holsonbake, T.B., Goeger, M.P., Bildfell, R., Dietary CLA alters yolk and tissue FA composition and hepatic histopathology of laying hens (2002) Lipids, 37, pp. 751-757Choi, J.S., Koh, I.U., Jung, M.H., Song, J., Effects of three different conjugated linoleic acid preparations on insulin signalling, fat oxidation and mitochondrial function in rats fed a high-fat diet (2007) Br J Nutr, 98, pp. 264-275DeLany, J.P., West, D.B., Changes in body composition with conjugated linoleic acid (2000) J Am Coll Nutr, 19, pp. 487S-493SEaley, K.N., El-Sohemy, A., Archer, M.C., Effects of dietary conjugated linoleic acid on the expression of uncoupling proteins in mice and rats (2002) Lipids, 37, pp. 853-861Echtay, K.S., Murphy, M.P., Smith, R.A., Talbot, D.A., Brand, M.D., Superoxide activates mitochondrial uncoupling protein 2 from the matrix side. Studies using targeted antioxidants (2002) J Biol Chem, 277, pp. 47129-47135Echtay, K.S., Roussel, D., St-Pierre, J., Jekabsons, M.B., Cadenas, S., Stuart, J.A., Harper, J.A., Brand, M.D., Superoxide activates mitochondrial uncoupling proteins (2002) Nature, 415, pp. 96-99Fruchart, J.C., Duriez, P., Staels, B., Peroxisome proliferatoractivated receptor-alpha activators regulate genes governing lipoprotein metabolism, vascular inflammation and atherosclerosis (1999) Curr Opin Lipidol, 10, pp. 245-257Garcia-Ruiz, C., Colell, A., Mari, M., Morales, A., Fernandez-Checa, J.C., Direct effect of ceramide on the mitochondrial electron transport chain leads to generation of reactive oxygen species. Role of mitochondrial glutathione (1997) J Biol Chem, 272, pp. 11369-11377Garlid, K.D., Jabůrek, M., Jezek, P., Varecha, M., How do uncoupling proteins uncouple? (2000) Biochim Biophys Acta, 1459, pp. 383-389Gavino, V.C., Gavino, G., Leblanc, M.J., Tuchweber, B., An isomeric mixture of conjugated linoleic acids but not pure cis-9, trans-11-octadecadienoic acid affects body weight gain and plasma lipids in hamsters (2000) J Nutr, 130, pp. 27-29Gholam, P.M., Flancbaum, L., Machan, J.T., Charney, D.A., Kotler, D.P., Nonalcoholic fatty liver disease in severely obese subjects (2007) Am J Gastroenterol, 102, pp. 399-408Griinari, J.M., Corl, B.A., Lacy, S.H., Chouinard, P.Y., Nurmela, K.V., Bauman, D.E., Conjugated linoleic acid is synthesized endogenously in lactating dairy cows by Delta (9)-desaturase (2000) J Nutr, 130, pp. 2285-2291Haddad, R., Sparrapan, R., Eberlin, M.N., Desorption sonic spray ionization for (high) voltage-free ambient mass spectrometry (2006) Rapid Commun Mass Spectrom, 20, pp. 2901-2905Haddad, R., Sparrapan, R., Kotiaho, T., Eberlin, M.N., Easy ambient sonic-spray ionization-membrane interface mass spectrometry for direct analysis of solution constituents (2008) Anal Chem, 80, pp. 898-903Halade, G.V., Rahman, M.M., Fernandes, G., Effect of CLA isomers and their mixture on aging C57Bl/6J mice (2009) Eur J Nutr, 48, pp. 409-418Hissin, P.J., Hilf, R., A fluorometric method for determination of oxidized and reduced glutathione in tissues (1976) Anal Biochem, 74, pp. 214-226Jaudszus, A., Moeckel, P., Hamelmann, E., Jahreis, G., Trans-10, cis-12-CLA-caused lipodystrophy is associated with profound changes of fatty acid profiles of liver, white adipose tissue and erythrocytes in mice: Possible link to tissue-specific alterations of fatty acid desaturation (2010) Ann Nutr Metab, 57, pp. 103-111Jezek, P., Garlid, K.D., Mammalian mitochondrial uncoupling proteins (1998) Int J Biochem Cell Biol, 30, pp. 1163-1168Kennedy, A., Martinez, K., Schmidt, S., Mandrup, S., LaPoint, K., McIntosh, M., Antiobesity mechanisms of action of conjugated linoleic acid (2010) J Nutr Biochem, 21, pp. 171-179Konig, B., Spielmann, J., Haase, K., Brandsch, C., Kluge, H., Stangl, G.I., Effects offish oil and conjugated linoleic acids on expression of target genes of PPAR alpha and sterol regulatory elementbinding proteins in the liver of laying hens (2008) Br J Nutr, 100, pp. 355-363Kritchevsky, D., Tepper, S.A., Wright, S., Tso, P., Czarnecki, S.K., Influence of conjugated linoleic acid (CLA) on establishment and progression of atherosclerosis in rabbits (2000) J Am Coll Nutr, 19, pp. 472S-477SLin, H., Boylston, T.D., Chang, M.J., Luedecke, L.O., Shultz, T.D., Survey of the conjugated linoleic acid contents of dairy products (1995) J Dairy Sci, 78, pp. 2358-2365Moya-Camarena, S.Y., Heuvel, J.P.V., Blanchard, S.G., Leesnitzer, L.A., Belury, M.A., Conjugated linoleic acid is a potent naturally occurring ligand and activator of PPARalpha (1999) J Lipid Res, 40, pp. 1426-1433Nicholls, D.G., The bioenergetics of brown adipose tissue mitochondria (1976) FEBS Lett, 61, pp. 103-110Ohnuki, K., Haramizu, S., Ishihara, K., Fushiki, T., Increased energy metabolism and suppressed body fat accumulation in mice by a low concentration of conjugated linoleic acid (2001) Biosci Biotechnol Biochem, 65, pp. 2200-2204Ohnuki, K., Haramizu, S., Oki, K., Ishihara, K., Fushiki, T., A single oral administration of conjugated linoleic acid enhanced energy metabolism in mice (2001) Lipids, 36, pp. 583-587Ostrowska, E., Muralitharan, M., Cross, R.F., Bauman, D.E., Dunshea, F.R., Dietary conjugated linoleic acids increase lean tissue and decrease fat deposition in growing pigs (1999) J Nutr, 129, pp. 2037-2042Park, Y., Albright, K.J., Liu, W., Storkson, J.M., Cook, M.E., Pariza, M.W., Effect of conjugated linoleic acid on body composition in mice (1997) Lipids, 32, pp. 853-858Peters, J.M., Park, Y., Gonzalez, F.J., Pariza, M.W., Influence of conjugated linoleic acid on body composition and target gene expression in peroxisome proliferator-activated receptor alphanull mice (2001) Biochim Biophys Acta, 1533, pp. 233-242Rahman, M.M., Bhattacharya, A., Banu, J., Fernandes, G., Conjugated linoleic acid protects against age-associated bone loss in C57BL/6 female mice (2007) J Nutr Biochem, 18, pp. 467-474Rakhshandehroo, M., Hooiveld, G., Müller, M., Kersten, S., Comparative analysis of gene regulation by the transcription factor PPARalpha between mouse and human (2009) PLoS One, 4, pp. e6796Reznick, A.Z., Packer, L., Oxidative damage to proteins: Spectrophotometric method for the carbonyl assay (1994) Methods Enzymol, 233, pp. 357-363Ribot, J., Portillo, M.P., Picó, C., Macarulla, M.T., Palou, A., Effects of trans-10, cis-12 conjugated linoleic acid on the expression of uncoupling proteins in hamsters fed an atherogenic diet (2007) Br J Nutr, 97, pp. 1074-1082Roche, H.M., Noone, E., Sewter, C., Mc, B.S., Savage, D., Gibney, M.J., O'Rahilly, S., Vidal-Puig, A.J., Isomer-dependent metabolic effects of conjugated linoleic acid: Insights from molecular markers sterol regulatory element-binding protein-1c and LXRalpha (2002) Diabetes, 51, pp. 2037-2044Ryder, J.W., Portocarrero, C.P., Song, X.M., Cui, L., Yu, M., Combatsiaris, T., Galuska, D., Houseknecht, K.L., Isomer-specific antidiabetic properties of conjugated linoleic acid. Improved glucose tolerance, skeletal muscle insulin action, and UCP-2 gene expression (2001) Diabetes, 50, pp. 1149-1157Samec, S., Seydoux, J., Dulloo, A.G., Role of UCP homologues in skeletal muscles and brown adipose tissue: Mediators of thermogenesis or regulators of lipids as fuel substrate? (1998) FASEB J, 12, pp. 715-724Schild, L., Reinheckel, T., Wiswedel, I., Augustin, W., Short-term impairment of energy production in isolated rat liver mitochondria by hypoxia/reoxygenation: Involvement of oxidative protein modification (1997) Biochem J, 15, pp. 205-210Schönfeld, P., Wojtczak, L., Fatty acids as modulators of the cellular production of reactive oxygen species (2008) Free Radic Biol Med, 45, pp. 231-241Schoonjans, K., Staels, B., Auwerx, J., Role of the peroxisome proliferator-activated receptor (PPAR) in mediating the effects of fibrates and fatty acids on gene expression (1996) J Lipid Res, 37, pp. 907-925Semighini, C.P., Marins, M., Goldman, M.H.S., Goldman, G.H., Quantitative analysis of the relative transcript levels of ABC transporter Atr genes in Aspergillus nidulans by real-time reverse transcripition-PCR assay (2002) Appl Environ Microbiol, 68, pp. 1351-1357Simas, R.C., Catharino, R.R., Cunha, I.B.S., Cabral, E.C., Barrera-Arellano, D., Eberlin, M.N., Instantaneous characterization of vegetable oils via TAG and FFA profiles by easy ambient sonicspray ionization mass spectrometry (2010) Analyst, 135, pp. 735-744Takahashi, Y., Kushiro, M., Shinohara, K., Ide, T., Dietary conjugated linoleic acid reduces body fat mass and affects gene expression of proteins regulating energy metabolism in mice (2002) Comp Biochem Physiol B Biochem Mol Biol, 133, pp. 395-404Terpstra, A.H., Effect of conjugated linoleic acid on body composition and plasma lipids in humans: An overview of the literature (2004) Am J Clin Nutr, 79, pp. 352-361Tsuboyama-Kasaoka, N., Takahashi, M., Tanemura, K., Kim, H.J., Tange, T., Okuyama, H., Conjugated linoleic acid supplementation reduces adipose tissue by apoptosis and develops lipodystrophy in mice (2000) Diabetes, 49, pp. 1534-1542Tsuboyama-Kasaoka, N., Miyazaki, H., Kasaoka, S., Ezaki, O., Increasing the amount of fat in a conjugated linoleic acid-supplemented diet reduces lipodystrophy in mice (2003) J Nutr, 133, pp. 1793-1799Wang, Y.W., Jones, P.J., Conjugated linoleic acid and obesity control: Efficacy and mechanisms (2004) Int J Obes Relat Metab Disord, 28, pp. 941-955West, D.B., Delany, J.P., Camet, P.M., Blohm, F., Truett, A.A., Scimeca, J., Effects of conjugated linoleic acid on body fat and energy metabolism in the mouse (1998) Am J Physiol, 275, pp. R667-R672West, D.B., Blohm, F.Y., Truett, A.A., DeLany, J.P., Conjugated linoleic acid persistently increases total energy expenditure in AKR/J mice without increasing uncoupling protein gene expression (2000) J Nutr, 130, pp. 2471-2477World Health Organization, Programmes and Projects, Nutrition Topics, Controlling the Global Obesity Epidemic, , http://www.who.int/nutrition/topics/obesity/en, Accessed April 5, 2012Zhou, M., Diwu, Z., Panchuk-Voloshina, N., Haugland, R.P., A stable nonfluorescent derivative of resorufin for the fluorometric determination of trace hydrogen peroxide: Applications in detecting the activity of phagocyte NADPH oxidase and other oxidases (1997) Anal Biochem, 15, pp. 162-16

    Visualizing Inhibition Of Fatty Acid Synthase Through Mass Spectrometric Analysis Of Mitochondria From Melanoma Cells

    No full text
    Fatty acid synthase (FASN) is the metabolic enzyme responsible for the endogenous synthesis of the saturated long-chain fatty acid palmitate. In contrast to most normal cells, FASN is overexpressed in a variety of human cancers including cutaneous melanoma, in which its levels of expression are associated with a poor prognosis and depth of invasion. Recently, we have demonstrated the mitochondrial involvement in FASN inhibition-induced apoptosis in melanoma cells. Herein we compare, via electrospray ionization mass spectrometry (ESI-MS), free fatty acids (FFA) composition of mitochondria isolated from control (EtOH-treated cells) and Orlistat-treated B16-F10 mouse melanoma cells. Principal component analysis (PCA) was applied to the ESI-MS data and found to separate the two groups of samples. Mitochondria from control cells showed predominance of six ions, that is, those of m/z 157 (Pelargonic, 9:0), 255 (Palmitic, 16:0), 281 (Oleic, 18:1), 311 (Arachidic, 20:0), 327 (Docosahexaenoic, 22:6) and 339 (Behenic, 22:0). In contrast, FASN inhibition with Orlistat changes significantly mitochondrial FFA composition by reducing synthesis of palmitic acid, and its elongation and unsaturation products, such as arachidic and behenic acids, and oleic acid, respectively. ESI-MS of mitochondria isolated from Orlistat-treated cells presented therefore three major ions of m/z 157 (Pelargonic, 9:0), 193 (unknown) and 199 (Lauric, 12:0). These findings demonstrate therefore that FASN inhibition by Orlistat induces significant changes in the FFA composition of mitochondria. © 2011 John Wiley & Sons, Ltd.253449452Smith, S., (1994) FASEB J, 8, p. 1248Menendez, J.A., Lupu, R., (2007) Nat. Rev. Cancer, 7, p. 763Weiss, L., Hoffmann, G.E., Schreiber, R., (1986) Biol. Chem. Hoppe Seyler, 367, p. 905Kuhajda, F.P., (2000) Nutrition, 16, p. 202Alo, P.L., Visca, P., Framarino, M.L., (2000) Oncol. Rep., 7, p. 1383Pizer, E.S., Jackisch, C., Wood, F.D., (1996) Cancer Res., 56, p. 2745Pizer, E.S., Wood, F.D., Heine, H.S., (1996) Cancer Res., 56, p. 1189Gansler, T.S., Hardman III, W., Hunt, D.A., (1997) Hum. Pathol., 28, p. 686Dhanasekaran, S.M., Barrette, T.R., Ghosh, D., (2001) Nature, 412, p. 822Swinnen, J.V., Roskams, T., Joniau, S., (2002) Int. J. Cancer, 98, p. 19Innocenzi, D., Alo, P.L., Balzani, A., (2003) J. Cutan. Pathol., 30, p. 23Rossi, S., Graner, E., Febbo, P., (2003) Mol. Cancer Res., 1, p. 707Takahiro, T., Shinichi, K., Toshimitsu, S., (2003) Clin. Cancer Res., 9, p. 2204Visca, P., Sebastiani, V., Botti, C., (2004) Anticancer Res., 24, p. 4169Kapur, P., Rakheja, D., Roy, L.C., (2005) Mod. Pathol., 18, p. 1107Van De Sande, T., Roskams, T., Lerut, E., (2005) J. Pathol., 206, p. 214Rossi, S., Ou, W., Tang, D., (2006) J. Pathol., 209, p. 369Dowling, S., Cox, J., Cenedella, R.J., (2009) Lipids, 44, p. 489Furuya, Y., Akimoto, S., Yasuda, K., (1997) Anticancer Res., 17, p. 4589Pizer, E.S., Chrest, F.J., Digiuseppe, J.A., (1998) Cancer Res., 58, p. 4611Li, J.N., Gorospe, M., Chrest, F.J., (2001) Cancer Res., 61, p. 1493Zhou, W., Han, W.F., Landree, L.E., (2007) Cancer Res., 67, p. 2964Kridel, S.J., Axelrod, F., Rozenkrantz, N., (2004) Cancer Res., 64, p. 2070Carvalho, M.A., Zecchin, K.G., Seguin, F., (2008) Int. J. Cancer, 123, p. 2557Zecchin, K.G., Rossato, F.A., Raposo, H.F., (2010) Lab. Invest., , DOI: 10.1038/labinvest.2010.157Swinnen, J.V., Van Veldhoven, P.P., Timmermans, L., (2003) Biochem. Biophys. Res. Commun., 302, p. 898Catharino, R.R., Haddad, R., Cabrini, L.G., (2005) Anal. Chem., 77, p. 7429Yang, L., Bennett, R., Strum, J., (2009) Anal. Bioanal. Chem., 393, p. 643Fenn, J.B., Mann, M., Meng, C.K., (1989) Science, 246, p. 64Catharino, R.R., Milagre, H.M.S., Saraiva, A.S., (2007) Energy Fuels, 21, p. 3698Ferreira, C.R., Souza, G., Riccio, M.F., (2009) Rapid Commun. Mass Spectrom., 23, p. 1313Santos, L.S., Catharino, R.R., Aguiar, C.L., (2006) J. Radioanal. Nucl. Chem., 269, p. 505Martin, E.B., Morris, A.J., Zhang, J., (1996) IEE Proc. - Control Theory and Applications, 143, p. 132Sumner, L.W., Mendes, P., Dixon, R.A., (2003) Phytochemistry, 62, p. 817Ferreira, C.R., Saraiva, A.S., Catharino, R.R., (2010) J. Lipid Res., , DOI: 10.1194/jlr.D001768Knowles, L.M., Axelrod, F., Browne, C.D., (2004) J. Biol. Chem., 279, p. 30540Abou-Khalil, S., Abou-Khalil, W.H., Planas, L., (1985) Biochem. Biophys. Res. Commun., 127, p. 1039Bligh, E.G., Dyer, W.J., (1959) Can. J. Biochem. Physiol., 37, p. 911Schrauwen, P., Saris, W.H.M., Hesselink, M.K.C., (2001) FASEB J, 15, p. 2497De Freitas, E.R.L., Soares, P.R.O., Santos, R.D., (2008) J. Nanosci. Nanotechnol., 8, p. 2385Liu, J., Shimizu, K., Kondo, R., (2009) Chem. Biodivers., 6, p. 50

    Mitochondrial Atp-sensitive K+ Channels As Redox Signals To Liver Mitochondria In Response To Hypertriglyceridemia

    No full text
    We have recently demonstrated that hypertriglyceridemic (HTG) mice present both elevated body metabolic rates and mild mitochondrial uncoupling in the liver owing to stimulated activity of the ATP-sensitive potassium channel (mitoKATP). Because lipid excess normally leads to cell redox imbalance, we examined the hepatic oxidative status in this model. Cell redox imbalance was evidenced by increased total levels of carbonylated proteins, malondialdehydes, and GSSG/GSH ratios in HTG livers compared to wild type. In addition, the activities of the extramitochondrial enzymes NADPH oxidase and xanthine oxidase were elevated in HTG livers. In contrast, Mn-superoxide dismutase activity and content, a mitochondrial matrix marker, were significantly decreased in HTG livers. Isolated HTG liver mitochondria presented lower rates of H2O2 production, which were reversed by mitoKATP antagonists. In vivo antioxidant treatment with N-acetylcysteine decreased both mitoKATP activity and metabolic rates in HTG mice. These data indicate that high levels of triglycerides increase reactive oxygen generation by extramitochondrial enzymes that promote mitoKATP activation. The mild uncoupling mediated by mitoKATP increases metabolic rates and protects mitochondria against oxidative damage. Therefore, a biological role for mitoKATP as a redox sensor is shown here for the first time in an in vivo model of systemic and cellular lipid excess. © 2009 Elsevier Inc. All rights reserved.471014321439Grundy, S.M., Brewer Jr., H.B., Cleeman, J.I., Smith Jr., S.C., Lenfant, C., Definition of metabolic syndrome: report of the National Heart, Lung, and Blood Institute/American Heart Association Conference on Scientific Issues Related to Definition (2004) Circulation, 109, pp. 433-438Sarwar, N., Danesh, J., Eiriksdottir, G., Sigurdsson, G., Wareham, N., Bingham, S., Boekholdt, S.M., Gudnason, V., Triglycerides and the risk of coronary heart disease: 10,158 incident cases among 262,525 participants in 29 western prospective studies (2007) Circulation, 115, pp. 450-458Griendling, K.K., FitzGerald, G.A., Oxidative stress and cardiovascular injury. Part II. Animal and human studies (2003) Circulation, 108, pp. 2034-2040Oliveira, H.C., Cosso, R.G., Alberici, L.C., Maciel, E.N., Salerno, A.G., Dorighello, G.G., Velho, J.A., Vercesi, A.E., Oxidative stress in atherosclerosis-prone mouse is due to low antioxidant capacity of mitochondria (2005) FASEB J., 19, pp. 278-280Chisolm, G.M., Steinberg, D., The oxidative modification hypothesis of atherogenesis: an overview (2000) Free Radic. Biol. Med., 28, pp. 1815-1826Baynes, J.W., Role of oxidative stress in development of complications in diabetes (1991) Diabetes, 40, pp. 405-412Ford, E.S., Will, J.C., Bowman, B.A., Narayan, K.M., Diabetes mellitus and serum carotenoids: findings from the Third National Health and Nutrition Examination Survey (1999) Am. J. Epidemiol., 149, pp. 168-176Saxena, R., Madhu, S.V., Shukla, R., Prabhu, K.M., Gambhir, J.K., Postprandial hypertriglyceridemia and oxidative stress in patients of type 2 diabetes mellitus with macrovascular complications (2005) Clin. Chim. Acta, 359, pp. 101-108Cardona, F., Túnez, I., Tasset, I., Garrido-Sánchez, L., Collantes, E., Tinahones, F.J., Circulating antioxidant defences are decreased in healthy people after a high-fat meal (2008) Br. J. Nutr., 100, pp. 312-316Diniz, Y.S., Rocha, K.K., Souza, G.A., Galhardi, C.M., Ebaid, G.M., Rodrigues, H.G., Novelli Filho, J.L., Novelli, E.L., Effects of N-acetylcysteine on sucrose-rich diet-induced hyperglycaemia, dyslipidemia and oxidative stress in rats (2006) Eur. J. Pharmacol., 543, pp. 151-157Cardona, F., Tunez, I., Tasset, I., Murri, M., Tinahones, F.J., Similar increase in oxidative stress after fat overload in persons with baseline hypertriglyceridemia with or without the metabolic syndrome (2008) Clin. Biochem., 41, pp. 701-705Schönfeld, P., Wojtczak, L., Fatty acids as modulators of the cellular production of reactive oxygen species (2008) Free Radic. Biol. Med., 45, pp. 231-241Stocker, R., Keaney Jr., J.F., Role of oxidative modifications in atherosclerosis (2004) Physiol. Rev., 84, pp. 1381-1478Hiramatsu, K., Arimori, S., Increased superoxide production by mononuclear cells of patients with hypertriglyceridemia and diabetes (1998) Diabetes, 37, pp. 832-837Prónai, L., Hiramatsu, K., Saigusa, Y., Nakazawa, H., Low superoxide scavenging activity associated with enhanced superoxide generation by monocytes from male hypertriglyceridemia with and without diabetes (1991) Atherosclerosis, 90, pp. 39-47Roberts, C.K., Barnard, R.J., Sindhu, R.K., Jurczak, M., Ehdaie, A., Vaziri, N.D., Oxidative stress and dysregulation of NAD(P)H oxidase and antioxidant enzymes in diet-induced metabolic syndrome (2006) Metabolism, 55, pp. 928-934Furukawa, S., Fujita, T., Shimabukuro, M., Iwaki, M., Yamada, Y., Nakajima, Y., Nakayama, O., Shimomura, I., Increased oxidative stress in obesity and its impact on metabolic syndrome (2004) J. Clin. Invest., 114, pp. 1752-1761Alberici, L.C., Oliveira, H.C., Bighetti, E.J., de Faria, E.C., Degaspari, G.R., Souza, C.T., Vercesi, A.E., Hypertriglyceridemia increases mitochondrial resting respiration and susceptibility to permeability transition (2003) J. Bioenerg. Biomembr., 35, pp. 451-457Alberici, L.C., Oliveira, H.C., Patrício, P.R., Kowaltowski, A.J., Vercesi, A.E., Hyperlipidemic mice present enhanced catabolism and higher mitochondrial ATP-sensitive K+channel activity (2006) Gastroenterology, 131, pp. 1228-1234Garlid, K.D., Paucek, P., Mitochondrial potassium transport: the K(+) cycle (2003) Biochim. Biophys. Acta, 1606, pp. 23-41Boveris, A., Mitochondrial production of superoxide radical and hydrogen peroxide (1977) Adv. Exp. Med. Biol., 78, pp. 67-82Skulachev, V.P., Uncoupling: new approaches to an old problem of bioenergetics (1998) Biochim. Biophys. Acta, 1363, pp. 100-124Facundo, H.T., de Paula, J.G., Kowaltowski, A.J., Mitochondrial ATP-sensitive K+channels are redox-sensitive pathways that control reactive oxygen species production (2007) Free Radic. Biol. Med., 42, pp. 1039-1048Bligh, E.G., Dyer, W.J.A., rapid method of total lipid extraction and purification (1959) Can. J. Biochem. Physiol., 37, pp. 911-917Iossa, S., Lionetti, L., Mollica, M.P., Barletta, A., Liverini, G., Oxidative activity in mitochondria isolated from rat liver at different stages of development (1998) Cell Biochem. Funct., 16, pp. 261-268Zhou, M., Diwu, Z., Panchuk-Voloshina, N., Haugland, R.P., A stable nonfluorescent derivative of resorufin for the fluorometric determination of trace hydrogen peroxide: applications in detecting the activity of phagocyte NADPH oxidase and other oxidases (1997) Anal. Biochem., 15, pp. 162-168Beauchamp, C.O., Fridovich, I., Isozymes of superoxide dismutase from wheat germ (1973) Biochim. Biophys. Acta, 317, pp. 50-64Morton, R.L., Ikle, D., White, C.W., Loss of lung mitochondrial aconitase activity due to hyperoxia in bronchopulmonary dysplasia in primates (1998) Am. J. Physiol., 274, pp. 127-133Racker, E., Spectrophotometric measurements of the enzymatic formation of fumaric and cis-aconitic acids (1950) Biochim. Biophys. Acta, 4, pp. 211-214Stirpe, F., Della Corte, E., The regulation of rat liver xanthine oxidase: conversion of type D (dehydrogenase) into type O (oxidase) by a thermolabile factor, and reversibility by dithioerythritol (1970) Biochim. Biophys. Acta, 212, pp. 195-197Oliveira, C.P., Alves, V.A., Lima, V.M., Stefano, J.T., Debbas, V., Sá, S.V., Wakamatsu, A., Carrilho, F.J., Modulation of hepatic microsomal triglyceride transfer protein (MTP) induced by S-nitroso-N-acetylcysteine in ob/ob mice (2007) Biochem. Pharmacol., 74, pp. 290-297Hissin, P.J., Hilf, R.A., fluorometric method for determination of oxidized and reduced glutathione in tissues (1976) Anal. Biochem., 74, pp. 214-226Reznick, A.Z., Packer, L., Oxidative damage to proteins: spectrophotometric method for the carbonyl assay (1994) Methods Enzymol., 233, pp. 357-363Schild, L., Reinheckel, T., Wiswedel, I., Augustin, W., Short-term impairment of energy production in isolated rat liver mitochondria by hypoxia/reoxygenation: involvement of oxidative protein modification (1997) Biochem. J., 15, pp. 205-210Calegario, F.F., Cosso, R.G., Fagian, M.M., Almeida, F.V., Jardim, W.F., Jezek, P., Arruda, P., Vercesi, A.E., Stimulation of potato tuber respiration by cold stress is associated with an increased capacity of both plant uncoupling mitochondrial protein (PUMP) and alternative oxidase (2003) J. Bioenerg. Biomembr., 35, pp. 211-220McGarry, J.D., Foster, D.W., Regulation of hepatic fatty acid oxidation and ketone body production. (1980) Annu. Rev. Biochem., 49, pp. 395-420Gardner, P.R., Raineri, I., Epstein, L.B., White, C.W., Superoxide radical and iron modulate aconitase activity in mammalian cells (1995) J. Biol. Chem., 270, pp. 13399-13405Teli, M.R., James, O.F., Burt, A.D., Bennett, M.K., Day, C.P., The natural history of nonalcoholic fatty liver (1995) Hepatology, 22, pp. 1714-1719Yamaguchi, K., Yang, L., McCall, S., Huang, J., Yu, X.X., Pandey, S.K., Bhanot, S., Diehl, A.M., Inhibiting triglyceride synthesis improves hepatic steatosis but exacerbates liver damage and fibrosis in obese mice with nonalcoholic steatohepatitis (2007) Hepatology, 45, pp. 1366-1374Leclercq, I.A., Farrell, G.C., Field, J., Bell, D.R., Gonzalez, F.J., Robertson, G.R., CYP2E1 and CYP4A as microsomal catalysts of lipid peroxides in murine nonalcoholic steatohepatitis (2000) J. Clin. Invest., 105, pp. 1067-1075Cighetti, G., Bortone, L., Sala, S., Allevi, P., Mechanisms of action of malondialdehyde and 4-hydroxynonenal on xanthine oxidoreductase (2001) Arch. Biochem. Biophys., 389, pp. 195-200Pégorier, J.P., Le May, C., Girard, J., Control of gene expression by fatty acids (2004) J. Nutr., 134, pp. 2444S-2449SBakker, S.J., Ijzerman, R.G., Teerlink, T., Westerhoff, H.V., Gans, R.O., Heine, R.J., Cytosolic triglycerides and oxidative stress in central obesity: the missing link between excessive atherosclerosis, endothelial dysfunction, and beta-cell failure? (2000) Atherosclerosis, 148, pp. 17-21Meilhac, O., Zhou, M., Santanam, N., Parthasarathy, S., Lipid peroxides induce expression of catalase in cultured vascular cells (2000) J. Lipid. Res., 41, pp. 1205-1213Kao, P.F., Lee, W.S., Liu, J.C., Chan, P., Tsai, J.C., Hsu, Y.H., Chang, W.Y., Liao, S.S., Downregulation of superoxide dismutase activity and gene expression in cultured rat brain astrocytes after incubation with vitamin C (2003) Pharmacology, 69, pp. 1-6Povoa Jr., H., Sá, L.D., Lessa, V.M., Xanthine oxidase and triglycerides in serum of patients with hyperlipoproteinemia, type IV (1984) Biomed. Biochim. Acta, 43, pp. 1201-1203Schröder, K., Vecchione, C., Jung, O., Schreiber, J.G., Shiri-Sverdlov, R., van Gorp, P.J., Busse, R., Brandes, R.P., Xanthine oxidase inhibitor tungsten prevents the development of atherosclerosis in ApoE knockout mice fed a Western-type diet (2006) Free Radic. Biol. Med., 41, pp. 1353-1360Brandes, R.P., Schröder, K., Differential vascular functions of Nox family NADPH oxidases (2008) Curr. Opin. Lipidol., 19, pp. 513-518Hsich, E., Segal, B.H., Pagano, P.J., Rey, F.E., Paigen, B., Deleonardis, J., Hoyt, R.F., Finkel, T., Vascular effects following homozygous disruption of p47(phox): an essential component of NADPH oxidase (2000) Circulation, 101, pp. 1234-1236Csont, T., Bereczki, E., Bencsik, P., Fodor, G., Görbe, A., Zvara, A., Csonka, C., Ferdinandy, P., Hypercholesterolemia increases myocardial oxidative and nitrosative stress thereby leading to cardiac dysfunction in apoB-100 transgenic mice (2007) Cardiovasc. Res., 76, pp. 100-109Wang, L., Sapuri-Butti, A.R., Aung, H.H., Parikh, A.N., Rutledge, J.C., Triglyceride-rich lipoprotein lipolysis increases aggregation of endothelial cell membrane microdomains and produces reactive oxygen species (2008) Am. J. Physiol. Heart Circ. Physiol., 295, pp. H237-244Zhang, D.X., Chen, Y.F., Campbell, W.B., Zou, A.P., Gross, G.J., Li, P.L., Characteristics and superoxide-induced activation of reconstituted myocardial mitochondrial ATP-sensitive potassium channels (2001) Circ. Res., 89, pp. 1177-1183Fornazari, M., de Paula, J.G., Castilho, R.F., Kowaltowski, A.J., Redox properties of the adenoside triphosphate-sensitive K+channel in brain mitochondria (2008) J. Neurosci. Res., 86, pp. 1548-1556Anderson, M.E., Glutathione: an overview of biosynthesis and modulation (1998) Chem. Biol. Interact., 111, pp. 1-1

    Inhibition of fatty acid synthase in melanoma cells activates the intrinsic pathway of apoptosis

    No full text
    Fatty acid synthase (FASN) is the metabolic enzyme responsible for the endogenous synthesis of the saturated long-chain fatty acid, palmitate. In contrast to most normal cells, FASN is overexpressed in a variety of human cancers, including cutaneous melanoma, in which its levels of expression are associated with tumor invasion and poor prognosis. We have previously shown that FASN inhibition with orlistat significantly reduces the number of spontaneous mediastinal lymph node metastases following the implantation of B16-F10 mouse melanoma cells in the peritoneal cavity of C57BL/6 mice. In this study, we investigate the biological mechanisms responsible for the FASN inhibition-induced apoptosis in B16-F10 cells. Both FASN inhibitors, cerulenin and orlistat, significantly reduced melanoma cell proliferation and activated the intrinsic pathway of apoptosis, as demonstrated by the cytochrome c release and caspase-9 and-3 activation. Further, apoptosis was preceded by an increase in both reactive oxygen species production and cytosolic calcium concentrations and independent of p53 activation and mitochondrial permeability transition. Taken together, these findings demonstrate the mitochondrial involvement in FASN inhibition-induced apoptosis in melanoma cells.912232240CONSELHO NACIONAL DE DESENVOLVIMENTO CIENTÍFICO E TECNOLÓGICO - CNPQCOORDENAÇÃO DE APERFEIÇOAMENTO DE PESSOAL DE NÍVEL SUPERIOR - CAPESFUNDAÇÃO DE AMPARO À PESQUISA DO ESTADO DE SÃO PAULO - FAPESP470539/2008-9Sem informação08/57471-7; 07/54639-

    Inhibition Of Fatty Acid Synthase In Melanoma Cells Activates The Intrinsic Pathway Of Apoptosis

    No full text
    Fatty acid synthase (FASN) is the metabolic enzyme responsible for the endogenous synthesis of the saturated long-chain fatty acid, palmitate. In contrast to most normal cells, FASN is overexpressed in a variety of human cancers, including cutaneous melanoma, in which its levels of expression are associated with tumor invasion and poor prognosis. We have previously shown that FASN inhibition with orlistat significantly reduces the number of spontaneous mediastinal lymph node metastases following the implantation of B16-F10 mouse melanoma cells in the peritoneal cavity of C57BL/6 mice. In this study, we investigate the biological mechanisms responsible for the FASN inhibition-induced apoptosis in B16-F10 cells. Both FASN inhibitors, cerulenin and orlistat, significantly reduced melanoma cell proliferation and activated the intrinsic pathway of apoptosis, as demonstrated by the cytochrome c release and caspase-9 and-3 activation. Further, apoptosis was preceded by an increase in both reactive oxygen species production and cytosolic calcium concentrations and independent of p53 activation and mitochondrial permeability transition. Taken together, these findings demonstrate the mitochondrial involvement in FASN inhibition-induced apoptosis in melanoma cells. © 2011 USCAP, Inc All rights reserved.912232240Smith, S., The animal fatty acid synthase: One gene, one polypeptide, seven enzymes (1994) FASEB J, 8, pp. 1248-1259Menendez, J.A., Lupu, R., Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis (2007) Nat Rev Cancer, 7, pp. 763-777Brink, J., Ludtke, S.J., Yang, C.Y., Quaternary structure of human fatty acid synthase by electron cryomicroscopy (2002) Proc Natl Acad Sci USA, 99, pp. 138-143Weiss, L., Hoffmann, G.E., Schreiber, R., Fatty-acid biosynthesis in man, a pathway of minor importance. Purification, optimal assay conditions, and organ distribution of fatty-acid synthase (1986) Biol Chem Hoppe Seyler, 367, pp. 905-912Kuhajda, F.P., Fatty-acid synthase and human cancer: New perspectives on its role in tumor biology (2000) Nutrition, 16, pp. 202-208Alo, P.L., Visca, P., Framarino, M.L., Immunohistochemical study of fatty acid synthase in ovarian neoplasms (2000) Oncol Rep, 7, pp. 1383-1388Pizer, E.S., Jackisch, C., Wood, F.D., Inhibition of fatty acid synthesis induces programmed cell death in human breast cancer cells (1996) Cancer Res, 56, pp. 2745-2747Pizer, E.S., Wood, F.D., Heine, H.S., Inhibition of fatty acid synthesis delays disease progression in a xenograft model of ovarian cancer (1996) Cancer Res, 56, pp. 1189-1193Gansler, T.S., Hardman Iii, W., Hunt, D.A., Increased expression of fatty acid synthase (OA-519) in ovarian neoplasms predicts shorter survival (1997) Hum Pathol, 28, pp. 686-692Dhanasekaran, S.M., Barrette, T.R., Ghosh, D., Delineation of prognostic biomarkers in prostate cancer (2001) Nature, 412, pp. 822-826Swinnen, J.V., Roskams, T., Joniau, S., Overexpression of fatty acid synthase is an early and common event in the development of prostate cancer (2002) Int J Cancer, 98, pp. 19-22Innocenzi, D., Alo, P.L., Balzani, A., Fatty acid synthase expression in melanoma (2003) J Cutan Pathol, 30, pp. 23-28Rossi, S., Graner, E., Febbo, P., Fatty acid synthase expression defines distinct molecular signatures in prostate cancer (2003) Mol Cancer Res, 1, pp. 707-715Takahiro, T., Shinichi, K., Toshimitsu, S., Expression of fatty acid synthase as a prognostic indicator in soft tissue sarcomas (2003) Clin Cancer Res, 9, pp. 2204-2212Visca, P., Sebastiani, V., Botti, C., Fatty acid synthase (FAS) is a marker of increased risk of recurrence in lung carcinoma (2004) Anticancer Res, 24, pp. 4169-4173Kapur, P., Rakheja, D., Roy, L.C., Fatty acid synthase expression in cutaneous melanocytic neoplasms (2005) Mod Pathol, 18, pp. 1107-1112Van De Sande, T., Roskams, T., Lerut, E., High-level expression of fatty acid synthase in human prostate cancer tissues is linked to activation and nuclear localization of Akt/PKB (2005) J Pathol, 206, pp. 214-219Rossi, S., Ou, W., Tang, D., Gastrointestinal stromal tumours overexpress fatty acid synthase (2006) J Pathol, 209, pp. 369-375Dowling, S., Cox, J., Cenedella, R.J., Inhibition of fatty acid synthase by Orlistat accelerates gastric tumor cell apoptosis in culture and increases survival rates in gastric tumor bearing mice in vivo (2009) Lipids, 44, pp. 489-498Furuya, Y., Akimoto, S., Yasuda, K., Apoptosis of androgenindependent prostate cell line induced by inhibition of fatty acid synthesis (1997) Anticancer Res, 17, pp. 4589-4593Pizer, E.S., Chrest, F.J., Digiuseppe, J.A., Pharmacological inhibitors of mammalian fatty acid synthase suppress DNA replication and induce apoptosis in tumor cell lines (1998) Cancer Res, 58, pp. 4611-4615Li, J.N., Gorospe, M., Chrest, F.J., Pharmacological inhibition of fatty acid synthase activity produces both cytostatic and cytotoxic effects modulated by p53 (2001) Cancer Res, 61, pp. 1493-1499Zhou, W., Han, W.F., Landree, L.E., Fatty acid synthase inhibition activates AMP-activated protein kinase in SKOV3 human ovarian cancer cells (2007) Cancer Res, 67, pp. 2964-2971Kridel, S.J., Axelrod, F., Rozenkrantz, N., Orlistat is a novel inhibitor of fatty acid synthase with antitumor activity (2004) Cancer Res, 64, pp. 2070-2075Alli, P.M., Pinn, M.L., Jaffee, E.M., Fatty acid synthase inhibitors are chemopreventive for mammary cancer in neu-N transgenic mice (2005) Oncogene, 24, pp. 39-46Bandyopadhyay, S., Zhan, R., Wang, Y., Mechanism of apoptosis induced by the inhibition of fatty acid synthase in breast cancer cells (2006) Cancer Res, 66, pp. 5934-5940Heiligtag, S.J., Bredehorst, R., David, K.A., Key role of mitochondria in cerulenin-mediated apoptosis (2002) Cell Death Differ, 9, pp. 1017-1025Liu, X., Shi, Y., Giranda, V.L., Inhibition of the phosphatidylinositol 3-kinase/Akt pathway sensitizes MDA-MB468 human breast cancer cells to cerulenin-induced apoptosis (2006) Mol Cancer Ther, 5, pp. 494-501Carvalho, M.A., Zecchin, K.G., Seguin, F., Fatty acid synthase inhibition with Orlistat promotes apoptosis and reduces cell growth and lymph node metastasis in a mouse melanoma model (2008) Int J Cancer, 123, pp. 2557-2565Knowles, L.M., Axelrod, F., Browne, C.D., A fatty acid synthase blockade induces tumor cell-cycle arrest by down-regulating Skp2 (2004) J Biol Chem, 279, pp. 30540-30545Chen, Y., McMillan-Ward, E., Kong, J., Oxidative stress induces autophagic cell death independent of apoptosis in transformed and cancer cells (2008) Cell Death Differ, 15, pp. 171-182Lebel, C.P., Ischiropoulos, H., Bondy, S.C., Evaluation of the probe 20 , 70-dichlorofluorescin as an indicator of reactive oxygen species formation and oxidative stress (1992) Chem Res Toxicol, 5, pp. 227-231Payne, C.M., Weber, C., Crowley-Skillicorn, C., Deoxycholate induces mitochondrial oxidative stress and activates NF-kappaB through multiple mechanisms in HCT-116 colon epithelial cells (2007) Carcinogenesis, 28, pp. 215-222Zecchin, K.G., Seidinger, A.L., Chiaratti, M.R., High Bcl-2/Bax ratio in Walker tumor cells protects mitochondria but does not prevent H2O2- induced apoptosis via calcineurin pathways (2007) J Bioenerg Biomembr, 39, pp. 186-194Tsien, R.Y., Fluorescence measurement and photochemical manipulation of cytosolic free calcium (1988) Trends Neurosci, 11, pp. 419-424Abou-Khalil, S., Abou-Khalil, W.H., Planas, L., Interaction of rhodamine 123 with mitochondria isolated from drug-sensitive and -resistant Friend leukemia cells (1985) Biochem Biophys Res Commun, 127, pp. 1039-1044Shepherd, D., Garland, P.B., ATP controlled acetoacetate and citrate synthesis by rat liver mitochondria oxidising palmitoyl-carnitine, and the inhibition of citrate synthase by ATP (1966) Biochem Biophys Res Commun, 22, pp. 89-93Campos, C.B., Paim, B.A., Cosso, R.G., Method for monitoring of mitochondrial cytochrome c release during cell death: Immunodetection of cytochrome c by flow cytometry after selective permeabilization of the plasma membrane (2006) Cytometry Part A, 69, pp. 515-523Green, D.R., Reed, J.C., Mitochondria and apoptosis (1998) Science, 281, pp. 1309-1312Garrido, C., Galluzzi, L., Brunet, M., Mechanisms of cytochrome c release from mitochondria (2006) Cell Death Differ, 13, pp. 1423-1433Ho, T.S., Ho, Y.P., Wong, W.Y., Fatty acid synthase inhibitors cerulenin and C75 retard growth and induce caspase-dependent apoptosis in human melanoma A-375 cells (2007) Biomed Pharmacother, 61, pp. 578-587Menendez, J.A., Vellon, L., Lupu, R., Antitumoral actions of the anti-obesity drug orlistat (XenicalTM) in breast cancer cells: Blockade of cell cycle progression, promotion of apoptotic cell death and PEA3-mediated transcriptional repression of Her2/neu (erbB-2) oncogene (2005) Ann Oncol, 16, pp. 1253-1267Menendez, J.A., Vellon, L., Lupu, R., Orlistat: From antiobesity drug to anticancer agent in Her-2/neu (erbB-2)-overexpressing gastrointestinal tumors? (2005) Exp Biol Med (Maywood), 230, pp. 151-154Kroemer, G., Galluzzi, L., Vandenabeele, P., Classification of cell death: Recommendations of the Nomenclature Committee on Cell Death 2009 (2009) Cell Death Differ, 16, pp. 3-11Straub, S.G., Sharp, G.W.G., Inhibition of insulin secretion by cerulenin might be due to impaired glucose metabolism (2007) Diabetes Metab Res Rev, 23, pp. 146-151Chajes, V., Cambot, M., Moreau, K., Acetyl-CoA carboxylase alpha is essential to breast cancer cell survival (2006) Cancer Res, 66, pp. 5287-5294Migita, T., Ruiz, S., Fornari, A., Fatty acid synthase: A metabolic enzyme and candidate oncogene in prostate cancer (2009) J Natl Cancer Inst, 101, pp. 519-532Lotem, J., Kama, R., Sachs, L., Suppression or induction of apoptosis by opposing pathways downstream from calcium-activated calcineurin (1999) Proc Natl Acad Sci USA, 96, pp. 12016-12020Wang, H.G., Pathan, N., Ethell, I.M., Ca2+-induced apoptosis through calcineurin dephosphorylation of BAD (1999) Science, 284, pp. 339-343Kowaltowski, A.J., Castilho, R.F., Vercesi, A.E., Mitochondrial permeability transition and oxidative stress (2001) FEBS Lett, 495, pp. 12-15Pizer, E.S., Thupari, J., Han, W.F., Malonyl-coenzyme-A is a potential mediator of cytotoxicity induced by fatty-acid synthase inhibition in human breast cancer cells and xenografts (2000) Cancer Res, 60, pp. 213-218Thupari, J.N., Pinn, M.L., Kuhajda, F.P., Fatty acid synthase inhibition in human breast cancer cells leads to malonyl-CoA-induced inhibition of fatty acid oxidation and cytotoxicity (2001) Biochem Biophys Res Commun, 285, pp. 217-223Zhang, Y., Guo, C., Yu, G., A pilot study of fatty acid metabolism in oral squamous cell carcinoma (2005) Int J Oral Maxillofac Surg, 34, pp. 78-81Paumen, M.B., Ishida, Y., Han, H., Direct interaction of the mitochondrial membrane protein carnitine palmitoyltransferase i with Bcl-2 (1997) Biochem Biophys Res Commun, 231, pp. 523-525Uddin, S., Siraj, A.K., Al-Rasheed, M., Fatty acid synthase and AKT pathway signaling in a subset of papillary thyroid cancers (2008) J Clin Endocrinol Metab, 93, pp. 4088-4097Mo, H., Elson, C.E., Apoptosis and cell-cycle arrest in human and murine tumor cells are initiated by isoprenoids (1999) J Nutr, 129, pp. 804-81
    corecore