The ketogenic diet reverses gene expression patterns and reduces reactive oxygen species levels when used as an adjuvant therapy for glioma

Phillip Stafford, Mohammed G Abdelwahab, Do Young Kim, Mark C Preul, Jong M Rho, Adrienne C Scheck, Phillip Stafford, Mohammed G Abdelwahab, Do Young Kim, Mark C Preul, Jong M Rho, Adrienne C Scheck

Abstract

Background: Malignant brain tumors affect people of all ages and are the second leading cause of cancer deaths in children. While current treatments are effective and improve survival, there remains a substantial need for more efficacious therapeutic modalities. The ketogenic diet (KD) - a high-fat, low-carbohydrate treatment for medically refractory epilepsy - has been suggested as an alternative strategy to inhibit tumor growth by altering intrinsic metabolism, especially by inducing glycopenia.

Methods: Here, we examined the effects of an experimental KD on a mouse model of glioma, and compared patterns of gene expression in tumors vs. normal brain from animals fed either a KD or a standard diet.

Results: Animals received intracranial injections of bioluminescent GL261-luc cells and tumor growth was followed in vivo. KD treatment significantly reduced the rate of tumor growth and prolonged survival. Further, the KD reduced reactive oxygen species (ROS) production in tumor cells. Gene expression profiling demonstrated that the KD induces an overall reversion to expression patterns seen in non-tumor specimens. Notably, genes involved in modulating ROS levels and oxidative stress were altered, including those encoding cyclooxygenase 2, glutathione peroxidases 3 and 7, and periredoxin 4.

Conclusions: Our data demonstrate that the KD improves survivability in our mouse model of glioma, and suggests that the mechanisms accounting for this protective effect likely involve complex alterations in cellular metabolism beyond simply a reduction in glucose.

Figures

Figure 1
Figure 1
MRI demonstrates the presence of tumors. MRI scans were done on tumor-bearing mice on days 9 and 15. Animals were positioned onto a probe in a head first, prone position. A T2 weighted MRI at 4.7 Tesla was obtained on 16 coronal sections of the animal's frontal lobe at a slice thickness of 0.5 mm.
Figure 2
Figure 2
Animals fed the ketogenic diet showed an increase in blood BHB levels. There was a statistically significant difference by 2-way ANOVA (p = 0.0067) in blood BHB levels in animals fed KD versus SD. There was no difference in the results obtained at the 2 different time points. Five animals were used per condition and the results are shown as the mean ± SEM.
Figure 3
Figure 3
Animals fed a ketogenic diet had a statistically significant increase in survival following tumor implantation. The number in parentheses is the number of animals in each treatment group.
Figure 4
Figure 4
Tumor growth in animals fed a standard diet (black circle) or a ketogenic diet (blue square). Animals were randomized to a treatment arm on day 3 post-implantation. Bioluminescence (photon count) was measured every 3 days. Results are an average of 5 animals for each diet.
Figure 5
Figure 5
Ketones reduced ROS levels. (A) Cells were treated with a cocktail containing either 1 mM (total 2 mM ketones) or 5 mM each (total 10 mM ketones) BHB and AcA for 24 hr prior to analysis. ROS levels were analyzed using DCF as described in the methods. Untreated tumor cells had high levels of ROS, and the application of either 2 mM or 10 mM ketones resulted in a dose dependent statistically significant decrease in the DCF signal demonstrating a reduction of ROS. (* = p ≤ 0.05),(* * = p ≤ 0.001); (B) The presence of increased reactive oxygen species (ROS) in tumor and the surrounding area relative to normal tissue is shown in a mouse fed a standard diet. Dihydroethidium (DHE) was imaged using an excitation wavelength of 500 nm and an emission wavelength of 620 nm. Spectral unmixing was used to differentiate the DHE signal from autofluorescence (shown in purple). The tumor was visualized using bioluminescence (shown in green); (C) Photomicrograph of brain slices from animals fed standard diet (SD) or ketogenic diet (KD). Areas from the tumor core and invading front of the tumor are shown. DCF fluorescence was analyzed as described in the methods section. There is a statistically significant difference in the amount of ROS in tumor vs normal brain when animals fed a standard diet were compared to those fed the ketogenic diet. (* = p ≤ 0.05); N.S. = not significant.
Figure 6
Figure 6
The KD alters overall gene expression to more closely resemble that seen in normal brain. Eight microarrays were analyzed by 2-way ANOVA for interaction effects, using standard Bonferonni multiple testing correction. There were strong interactions between ketogenic diet and normal diet in non-tumor classes, especially in context of standard diet non-tumor. The trend is that many expression profiles in tumor mice on a ketogenic diet seem to trend back to a profile seen with mice living on a standard diet having no tumor.
Figure 7
Figure 7
Expression of genes with significant differential expression in animals fed a KD versus SD. Log2 expression levels of genes that have ≥2-fold expression in at least one condition ratio. 95% confidence intervals are shown.

References

    1. Ruggiero A, Cefalo G, Garre ML, Massimino M, Colosimo C, Attina G, Phase II trial of temozolomide in children with recurrent high-grade glioma. J Neurooncol. 2005. pp. 1–6.
    1. Seyfried TN, Sanderson TM, El-Abbadi MM, McGowan R, Mukherjee P. Role of glucose and ketone bodies in the metabolic control of experimental brain cancer. Br J Cancer. 2003;89:1375–1382. doi: 10.1038/sj.bjc.6601269.
    1. Nebeling LC, Miraldi F, Shurin SB, Lerner E. Effects of a ketogenic diet on tumor metabolism and nutritional status in pediatric oncology patients: two case reports. J Am Coll Nutr. 1995;14:202–208.
    1. Nebeling LC, Lerner E. Implementing a ketogenic diet based on medium-chain triglyceride oil in pediatric patients with cancer. J Am Diet Assoc. 1995;95:693–697. doi: 10.1016/S0002-8223(95)00189-1.
    1. Zuccoli G, Marcello N, Pisanello A, Servadei F, Vaccaro S, Mukherjee P. et al.Metabolic management of glioblastoma multiforme using standard therapy together with a restricted ketogenic diet: Case Report. Nutrition and Metabolism. 2010;7:33–53. doi: 10.1186/1743-7075-7-33.
    1. Gasior M, Rogawski MA, Hartman AL. Neuroprotective and disease-modifying effects of the ketogenic diet. Behav Pharmacol. 2006;17:431–439. doi: 10.1097/00008877-200609000-00009.
    1. Kim DY, Rho JM. The ketogenic diet and epilepsy. Curr Opin Clin Nutr Metab Care. 2008;11:113–120. doi: 10.1097/MCO.0b013e3282f44c06.
    1. Fruehauf JP, Meyskens FL Jr. Reactive oxygen species: a breath of life or death? Clin Cancer Res. 2007;13:789–794. doi: 10.1158/1078-0432.CCR-06-2082.
    1. Weinberg F, Chandel NS. Reactive oxygen species-dependent signaling regulates cancer. Cell Mol Life Sci. 2009;66:3663–3673. doi: 10.1007/s00018-009-0099-y.
    1. Newcomb EW, Demaria S, Lukyanov Y, Shao Y, Schnee T, Kawashima N. et al.The combination of ionizing radiation and peripheral vaccination produces long-term survival of mice bearing established invasive GL261 gliomas. Clin Cancer Res. 2006;12:4730–4737. doi: 10.1158/1078-0432.CCR-06-0593.
    1. Jouanneau E, Poujol D, Gulia S, Le M, Blay JY, Belin MF. et al.Dendritic cells are essential for priming but inefficient for boosting antitumour immune response in an orthotopic murine glioma model. Cancer Immunol Immunother. 2006;55:254–267. doi: 10.1007/s00262-005-0040-7.
    1. Rho JM, Kim DW, Robbins CA, Anderson GD, Schwartzkroin PA. Age-dependent differences in flurothyl seizure sensitivity in mice treated with a ketogenic diet. Epilepsy Res. 1999;37:233–240. doi: 10.1016/S0920-1211(99)00068-6.
    1. Rho JM, Sarnat HB, Sullivan PG, Robbins CA, Kim DW. Lack of long-term histopathologic changes in brain and skeletal muscle of mice treated with a ketogenic diet. J Child Neurol. 2004;19:555–557.
    1. Maalouf M, Sullivan PG, Davis L, Kim DY, Rho JM. Ketones inhibit mitochondrial production of reactive oxygen species production following glutamate excitotoxicity by increasing NADH oxidation. Neuroscience. 2007;145:256–264. doi: 10.1016/j.neuroscience.2006.11.065.
    1. Stafford P, Brun M. Three methods for optimization of cross-laboratory and cross-platform microarray expression data. Nucl Acids Res. 2007;35:e72. doi: 10.1093/nar/gkl1133.
    1. Newcomb EW, Tamasdan C, Entzminger Y, Arena E, Schnee T, Kim M. et al.Flavopiridol inhibits the growth of GL261 gliomas in vivo: implications for malignant glioma therapy. Cell Cycle. 2004;3:230–234.
    1. Newcomb EW, Lymberis SC, Lukyanov Y, Shao Y, Schnee T, Devitt M. et al.Radiation Sensitivity of GL261 Murine Glioma Model and Enhanced Radiation Response by Flavopiridol. Cell Cycle. 2006;5:93–99.
    1. Szatmari T, Lumniczky K, Desaknai S, Trajcevski S, Hidvegi EJ, Hamada H. et al.Detailed characterization of the mouse glioma 261 tumor model for experimental glioblastoma therapy. Cancer Sci. 2006;97:546–553. doi: 10.1111/j.1349-7006.2006.00208.x.
    1. Pyles RB, Chalk CL, Balko MG, Miller MA, Dyer CA. et al.A syngeneic mouse glioma model for study of glioblastoma therapy. J Neuropath Exp Neurol. 1999;58:54–60. doi: 10.1097/00005072-199901000-00007.
    1. Laughlin KM, Luo D, Liu C, Shaw G, Warrington KH, Qiu J. et al.Hematopoietic- and Neurologic-Expressed Sequence 1 Expression in the Murine GL261 and High-Grade Human Gliomas. Pathol Oncol Res. 2009;15:12253–12267. doi: 10.1007/s12253-008-9147-4.
    1. Seyfried TN, Mukherjee P. Targeting energy metabolism in brain cancer: review and hypothesis. Nutrition and Metabolism. 2005;2:30–38. doi: 10.1186/1743-7075-2-30.
    1. Mahoney LB, Denny CA, Seyfried TN. Caloric restriction in C57BL/6J mice mimics therapeutic fasting in humans. Lipids Health Dis. 2006;5:13. doi: 10.1186/1476-511X-5-13.
    1. Vamecq J, Vallee L, Lesage F, Gressens P, Stables JP. Antiepileptic popular ketogenic diet: emerging twists in an ancient story. Prog Neurobiol. 2005;75:1–28. doi: 10.1016/j.pneurobio.2004.11.003.
    1. Dekundy A, Pietraszek M, Schaefer D, Cenci MA, Danysz W. Effects of group I metabotropic glutamate receptors blockade in experimental models of Parkinson's disease. Brain Res Bull. 2006;69:318–326. doi: 10.1016/j.brainresbull.2005.12.009.
    1. Freemantle E, Vandal M, Tremblay-Mercier J, Tremblay S, Blachere JC, Begin ME. et al.Omega-3 fatty acids, energy substrates, and brain function during aging. Prostaglandins Leukot Essent Fatty Acids. 2006;75:213–220. doi: 10.1016/j.plefa.2006.05.011.
    1. Jabre MG, Bejjani BP. Treatment of Parkinson disease with diet-induced hyperketonemia: a feasibility study. Neurology. 2006;66:617. doi: 10.1212/01.wnl.0000216108.57529.b1.
    1. Liao Y, Takashima S, Zhao H, Asano Y, Shintani Y, Minamino T. et al.Control of plasma glucose with alpha-glucosidase inhibitor attenuates oxidative stress and slows the progression of heart failure in mice. Cardiovasc Res. 2006;70:107–116. doi: 10.1016/j.cardiores.2006.01.021.
    1. Prins ML, Fujima LS, Hovda DA. Age-dependent reduction of cortical contusion volume by ketones after traumatic brain injury. J Neurosci Res. 2005;82:413–420. doi: 10.1002/jnr.20633.
    1. Zhao Z, Lange DJ, Voustianiouk A, MacGrogan D, Ho L, Suh J. et al.A ketogenic diet as a potential novel therapeutic intervention in amyotrophic lateral sclerosis. BMC Neurosci. 2006;7:29. doi: 10.1186/1471-2202-7-29.
    1. Mukherjee P, El-Abbadi MM, Kasperzyk JL, Ranes MK, Seyfried TN. Dietary restriction reduces angiogenesis and growth in an orthotopic mouse brain tumour model. Br J Cancer. 2002;86:1615–1621. doi: 10.1038/sj.bjc.6600298.
    1. Mukherjee P, Abate LE, Seyfried TN. Antiangiogenic and proapoptotic effects of dietary restriction on experimental mouse and human brain tumors. Clin Cancer Res. 2004;10:5622–5629. doi: 10.1158/1078-0432.CCR-04-0308.
    1. Mukherjee P, Mulrooney TJ, Marsh J, Blair D, Chiles TC, Seyfried TN. Differential effects of energy stress on AMPK phosphorylation and apoptosis in experimental brain tumor and normal brain. Mol Cancer. 2008;7:37. doi: 10.1186/1476-4598-7-37.
    1. Ito H, Daido S, Kanzawa T, Kondo S, Kondo Y. Radiation-induced autophagy is associated with LC3 and its inhibition sensitizes malignant glioma cells. Int J Oncol. 2005;26:1401–1410.
    1. Tabernero A, Medina JM, Giaume C. Glucose metabolism and proliferation in glia: role of astrocytic gap junctions. J Neurochem. 2006;99:1049–1061. doi: 10.1111/j.1471-4159.2006.04088.x.
    1. Marsh J, Mukherjee P, Seyfried TN. Drug/diet synergy for managing malignant astrocytoma in mice: 2-deoxy-D-glucose and the restricted ketogenic diet. Nutr Metab (Lond) 2008;5:33. doi: 10.1186/1743-7075-5-33.
    1. Noh HS, Lee HP, Kim DW, Kang SS, Cho GJ, Rho JM. et al.A cDNA microarray analysis of gene expression profiles in rat hippocampus following a ketogenic diet. Brain Res Mol Brain Res. 2004;129:80–87.
    1. Bough KJ, Wetherington J, Hassel B, Pare JF, Gawryluk JW, Greene JG. et al.Mitochondrial biogenesis in the anticonvulsant mechanism of the ketogenic diet. Ann Neurol. 2006;60:223–235. doi: 10.1002/ana.20899.
    1. Kennedy AR, Pissios P, Otu H, Xue B, Asakura K, Furukawa N. et al.A high-fat, ketogenic diet induces a unique metabolic state in mice. Am J Physiol Endocrinol Metab. 2007;292:E1724–E1739. doi: 10.1152/ajpendo.00717.2006.
    1. Muise ES, Azzolina B, Kuo DW, El-Sherbeini M, Tan Y, Yuan X. et al.Adipose fibroblast growth factor 21 is up-regulated by peroxisome proliferator-activated receptor gamma and altered metabolic states. Mol Pharmacol. 2008;74:403–412. doi: 10.1124/mol.108.044826.
    1. Erol E, Kumar LS, Cline GW, Shulman GI, Kelly DP, Binas B. Liver fatty acid binding protein is required for high rates of hepatic fatty acid oxidation but not for the action of PPARalpha in fasting mice. FASEB J. 2004;18:347–349.
    1. Sullivan PG, Rippy NA, Dorenbos K, Concepcion RC, Agarwal AK, Rho JM. The ketogenic diet increases mitochondrial uncoupling protein levels and activity. Ann Neurol. 2004;55:576–580. doi: 10.1002/ana.20062.
    1. Aykin-Burns N, Ahmad IM, Zhu Y, Oberley LW, Spitz DR. Increased levels of superoxide and H2O2 mediate the differential susceptibility of cancer cells versus normal cells to glucose deprivation. Biochem J. 2009;418:29–37. doi: 10.1042/BJ20081258.
    1. Landriscina M, Maddalena F, Laudiero G, Esposito F. Adaptation to oxidative stress, chemoresistance, and cell survival. Antioxid Redox Signal. 2009;11:2701–2716. doi: 10.1089/ars.2009.2692.
    1. Semenza GL. Regulation of cancer cell metabolism by hypoxia-inducible factor 1. Semin Cancer Biol. 2009;19:12–16. doi: 10.1016/j.semcancer.2008.11.009.
    1. Trachootham D, Alexandre J, Huang P. Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Nat Rev Drug Discov. 2009;8:579–591. doi: 10.1038/nrd2803.
    1. Chen Y, McMillan-Ward E, Kong J, Israels SJ, Gibson SB. Oxidative stress induces autophagic cell death independent of apoptosis in transformed and cancer cells. Cell Death Differ. 2008;15:171–182. doi: 10.1038/sj.cdd.4402233.
    1. Pore N, Jiang Z, Shu HK, Bernhard E, Kao GD, Maity A. Akt1 activation can augment hypoxia-inducible factor-1alpha expression by increasing protein translation through a mammalian target of rapamycin-independent pathway. Mol Cancer Res. 2006;4:471–479. doi: 10.1158/1541-7786.MCR-05-0234.
    1. Giglio P, Levin V. Cyclooxygenase-2 inhibitors in glioma therapy. Am J Ther. 2004;11:141–143. doi: 10.1097/00045391-200403000-00009.
    1. Mattila S, Tuominen H, Koivukangas J, Stenback F. The terminal prostaglandin synthases mPGES-1, mPGES-2, and cPGES are all overexpressed in human gliomas. Neuropathology. 2009;29:156–165. doi: 10.1111/j.1440-1789.2008.00963.x.
    1. Schonthal AH. Antitumor properties of dimethyl-celecoxib, a derivative of celecoxib that does not inhibit cyclooxygenase-2: implications for glioma therapy. Neurosurg Focus. 2006;20:E21. doi: 10.3171/foc.2006.20.4.14.
    1. Sobolewski C, Cerella C, Dicato M, Ghibelli L, Diederich M. The role of cyclooxygenase-2 in cell proliferation and cell death in human malignancies. Int J Cell Biol. 2010. p. 215158. Epub@2010 Mar 17.
    1. Kim CK, Joe YA, Lee SK, Kim EK, O E, Kim HK. et al.Enhancement of anti-tumor activity by low-dose combination of the recombinant urokinase kringle domain and celecoxib in a glioma model. Cancer Lett. 2010;288:251–260. doi: 10.1016/j.canlet.2009.07.008.
    1. Utomo A, Jiang X, Furuta S, Yun J, Levin DS, Wang YC. et al.Identification of a novel putative non-selenocysteine containing phospholipid hydroperoxide glutathione peroxidase (NPGPx) essential for alleviating oxidative stress generated from polyunsaturated fatty acids in breast cancer cells. J Biol Chem. 2004;279:43522–43529. doi: 10.1074/jbc.M407141200.
    1. Okado-Matsumoto A, Matsumoto A, Fujii J, Taniguchi N. Peroxiredoxin IV is a secretable protein with heparin-binding properties under reduced conditions. Journal of Biochemistry. 2000;127:493–501.
    1. Ziegler DR, Ribeiro LC, Hagenn M, Siqueira IR, Araujo E, Torres IL. et al.Ketogenic diet increases glutathione peroxidase activity in rat hippocampus. Neurochem Res. 2003;28:1793–1797. doi: 10.1023/A:1026107405399.
    1. Li D, Chen XQ, Li WJ, Yang YH, Wang JZ, Yu AC. Cytoglobin up-regulated by hydrogen peroxide plays a protective role in oxidative stress. Neurochem Res. 2007;32:1375–1380. doi: 10.1007/s11064-007-9317-x.
    1. Shivapurkar N, Stastny V, Okumura N, Girard L, Xie Y, Prinsen C. et al.Cytoglobin, the newest member of the globin family, functions as a tumor suppressor gene. Cancer Res. 2008;68:7448–7456. doi: 10.1158/0008-5472.CAN-08-0565.
    1. Cheng G, Lambeth JD. NOXO1, Regulation of lipid binding, localization, and activation of Nox1 by the Phox homology (PX) domain. J Biol Chem. 2004;279:4737–4742. doi: 10.1074/jbc.M305968200.
    1. Polytarchou C, Pfau R, Hatziapostolou M, Tsichlis PN. The JmjC domain histone demethylase Ndy1 regulates redox homeostasis and protects cells from oxidative stress. Molecular and Cellular Biology. 2008;28:7451–7464. doi: 10.1128/MCB.00688-08.

Source: PubMed

3
購読する