Nuclear magnetic resonance spectroscopy reveals metabolic changes in living cardiomyocytes after low doses of ionizing radiation

  • Michalina Gramatyka
  • Agnieszka Skorupa
  • Maria Sokół

Abstract

Several lines of evidence indicate that exposure of heart to ionizing radiation increases the risk of cardiotoxicity manifested by heart dysfunction and cardiovascular diseases. It was initially believed that the heart is an organ relatively resistant to radiation. Currently, however, it is suspected that even low doses of radiation (< 2 Gy) may have a negative impact on the cardiovascular system. Cardiotoxicity of ionizing radiation is associated with metabolic changes observed in cardiac cells injured by radiation. In this study we used human cardiomyocytes as a model system, and studied their metabolic response to radiation using high resolution magic angle spinning nuclear magnetic resonance techniques (HR MAS NMR).

Cultured in vitro human cardiomyocytes were exposed to ionizing radiation and their survival was assessed by clonogenic assay. Changes in apoptosis intensity and cell cycle distribution after irradiation were measured as well. NMR spectra of cardiomyocytes were acquired using Bruker Avance 400 MHz spectrometer at a spinning rate of 3200 Hz. Survival of cardiomyocytes after NMR experiments was assessed by the Trypan blue exclusion assay.

Exposure of cardiomyocytes to small doses of ionizing radiation had no effect on cell proliferation potential and intensity of cell death. However, analysis of metabolic profiles revealed changes in lipids, threonine, glycine, choline, valine, isoleucine, glutamate, reduced glutathione and taurine metabolism. The results of this study show that ionizing radiation affects metabolic profiles of cardiomyocytes even at low doses, which potentially have no effect on cell viability.

References

Andreadou I, Papaefthimiou M, Zira A, Constantinou M, Sigala F, Skaltsounis AL, Tsantili-Kakoulidou A, Iliodromitis EK, Kremastinos DT, Mikros E (2009) Metabonomic identification of novel biomarkers in doxorubicin cardiotoxicity and protective effect of the natural antioxidant oleuropein. NMR Biomed. 22:585-92. doi: 10.1002/nbm.1370

Bansal A, Harris RA, DeGrado TR (2012) Choline phosphorylation and regulation of transcription of choline kinase α in hypoxia. J Lipid Res 53:149-57. doi: 10.1194/jlr.M021030

Barba I, Garcia-Dorado D (2012) Metabolomics in Cardiovascular Disease: Towards Clinical Application. In: Coronary Artery Disease – New Insights and Novel Approaches. InTech, pp 207-224.

Barba I, Jaimez-Auguets E, Rodriguez-Sinovas A, Garcia-Dorado D (2007) 1H NMR-based metabolomic identification of at-risk areas after myocardial infarction in swine. MAGMA 20:265-71

Bezabeh T, Mowat MR, Jarolim L, Greenberg AH, Smith IC (2001) Detection of drug-induced apoptosis and necrosis in human cervical carcinoma cells using 1H NMR spectroscopy. Cell Death Differ 8:219-24. doi: 10.1038/sj.cdd.4400802

Blankenberg FG, Katsikis PD, Storrs RW, Beaulieu C, Spielman D, Chen JY, Naumovski L, Tait JF (1997) Quantitative analysis of apoptotic cell death using proton nuclear magnetic resonance spectroscopy. Blood 89:3778-86.

Boerma M, Sridharan V, Mao XW, Nelson GA, Cheema AK, Koturbash I, Singh SP, Tackett AJ, Hauer-Jensen M (2016) Effects of ionizing radiation on the heart. Mutat Res 770:319-327. doi: 10.1016/j.mrrev.2016.07.003

Bouillon K, Haddy N, Delaloge S, Garbay JR, Garsi JP, Brindel P, MousannifA, Lê MG, Labbe M, Arriagada R, Jougla E, Chavaudra J, Diallo I, Rubino C, de Vathaire F (2011) Long-term cardiovascular mortality after radiotherapy for breast cancer. J Am Coll Cardiol 57:445-52. doi: 10.1016/j.jacc.2010.08.638

Carvajal K, Moreno-Sánchez R (2003) Heart Metabolic Disturbances in Cardiovascular Diseases. Arch Med Res 34:89-99. doi: 10.1016/S0188-4409(03)00004-3

Darby SC, Ewertz M, McGale P, Bennet AM, Blom-Goldman U, Brønnum D, Correa C, Cutter D, Gagliardi G, Gigante B, Jensen MB, Nisbet A, Peto R, Rahimi K, Taylor C, Hall P (2013) Risk of ischemic heart disease in women after radiotherapy for breast cancer. N Engl J Med 368:987-98. doi: 10.1056/NEJMoa1209825

Delp MD, Charvat JM, Limoli CL, Globus RK, Ghosh P (2016) Apollo Lunar Astronauts Show Higher Cardiovascular Disease Mortality: Possible Deep Space Radiation Effects on the Vascular Endothelium. Sci Rep 6:29901. doi: 10.1038/srep29901

DiPaola RS (2002) To arrest or not to G(2)-M Cell-cycle arrest : commentary re: A. K. Tyagi et al., Silibinin strongly synergizes human prostate carcinoma DU145 cells to doxorubicin-induced growth inhibition, G(2)-M arrest, and apoptosis. Clin. ancer res., 8: 3512-3519, 2002. Clin Cancer Res 8:3311-4.

Eckhart AD, Beebe K, Milburn M (2012) Metabolomics as a key integrator for "omic" advancement of personalized medicine and future therapies. Clin Transl Sci 5:285-288. doi: 10.1111/j.1752-8062.2011.00388.x

Evans KM, Bodmer J, Edwards B, Levins J, O'Meara A, Ruhotina M, Smith R, Delaney T, Hoffman-Contois R, Boccuzzo L et al (2015) An Exploratory Analysis of Public Awareness and Perception of Ionizing Radiation and Guide to Public Health Practice in Vermont. J Environ Public Health 2015:476495. doi: 10.1155/2015/476495

Faller KM, Lygate CA, Neubauer S, Schneider JE (2013) (1)H-MR spectroscopy for analysis of cardiac lipid and creatine metabolism. Heart Fail Rev 18:657-68.doi: 10.1007/s10741-012-9341-z

Friis MB, Friborg CR, Schneider L, Nielsen MB, Lambert IH, Christensen ST, Hoffmann EK (2005) Cell shrinkage as a signal to apoptosis in NIH 3T3 fibroblasts. J Physiol 567(Pt 2):427-43. doi: 10.1113/jphysiol.2005.087130

Goudarzi M, Mak TD, Chen C, Smilenov LB, Brenner DJ, Fornace AJ (2014) The effect of low dose rate on metabolomic response to radiation in mice. Radiat Environ Biophys 53:645-57.doi: 10.1007/s00411-014-0558-1

Grande S, Luciani AM, Rosi A, Cherubini R, Conzato M, Guidoni L, Viti V (2001) Radiation effects on soluble metabolites in cultured HeLa cells examined by 1H MRS: changes in concentration of glutathione and of lipid catabolites induced by gamma rays and proton beams. Int J Cancer. 96 Suppl:27-42. doi: 10.1002/ijc.10345

Gu Y, Mi W, Ge Y, Liu H, Fan Q, Han C, Yang J, Han F, Lu X, Yu W (2010) GlcNAcylation plays an essential role in breast cancer metastasis. Cancer Res 70:6344-51. doi: 10.1158/0008-5472.CAN-09-1887

Harris RA, Joshi M, Jeoung NH (2004) Mechanisms responsible for regulation of branched-chain amino acid catabolism. Biochem Biophys Res Commun 313:391-6.

Jang WG, Park JY, Lee J, Bang E, Kim SR, Lee EK, Yun HJ, Kang CM, Hwang GS (2016) Investigation of relative metabolic changes in the organs and plasma of rats exposed to X-ray radiation using HR-MAS (1)H NMR and solution (1)H NMR. NMR Biomed 29:507-18. doi: 10.1002/nbm.3485

Jong CJ, Azuma J, Schaffer S (2012) Mechanism underlying the antioxidant activity of taurine: prevention of mitochondrial oxidant production. Amino Acids 42:2223-32. doi: 10.1007/s00726-011-0962-7

Kell DB, Brown M, Davey HM, Dunn WB, Spasic I, Oliver SG (2005) Metabolic footprinting and systems biology: the medium is the message. Nat Rev Microbiol 3:557-65.

Khurana S, Hollingsworth A, Piche M, Venkataraman K, Kumar A, Ross GM, Tai TC (2014) Antiapoptotic actions of methyl gallate on neonatal rat cardiac myocytes exposed to H2O2. Oxid Med Cell Longev 2014:657512. doi: 10.1155/2014/657512

Kwon YK, Ha IJ, Bae HW, Jang WG, Yun HJ, Kim SR, Lee EK, Kang CM, Hwang GS (2014) Dose-dependent metabolic alterations in human cells exposed to gamma irradiation. PLoS One 9:e113573. doi: 10.1371/journal.pone.0113573

Ladeirinha AFF (2011) Establishment of primary cell cultures from lung tissue biopsies and study of the cellular metabolic responses to cisplatin and radiation exposure. Disseration, Universidade de Coimbra

Laiakis EC, Hyduke DR, Fornace AJ (2012) Comparison of mouse urinary metabolic profiles after exposure to the inflammatory stressors γ radiation and lipopolysaccharide. Radiat Res 177:187-99.

Lang F, Ritter M, Gamper N, Huber S, Fillon S, Tanneur V, Lepple-Wienhues A, Szabo I, Gulbins E (2000) Cell volume in the regulation of cell proliferation and apoptotic cell death. Cell Physiol Biochem 10:417-28.

Lin H, Zhang J, Gao P (2009) Silent myocardial ischemia is associated with altered plasma phospholipids. J Clin Lab Anal 23:45-50. doi: 10.1002/jcla.20288

Little MP, Azizova TV, Bazyka D, Bouffler SD, Cardis E, Chekin S, Chumak VV, Cucinotta FA, de Vathaire F, Hall P, Harrison JD, Hildebrandt G, Ivanov V, Kashcheev VV, Klymenko SV, Kreuzer M, Laurent O, Ozasa K, Schneider T, Tapio S, Taylor AM, Tzoulaki I, Vandoolaeghe WL, Wakeford R, Zablotska LB, Zhang W, Lipshultz SE (2012) Systematic review and meta-analysis of circulatory disease from exposure to low-level ionizing radiation and estimates of potential population mortality risks. Environ Health Perspect 120:1503-11. doi: 10.1289/ehp.1204982

Locasale JW (2013) Serine, glycine and one-carbon units: cancer metabolism in full circle. Nat Rev Cancer 13:572-83. doi: 10.1038/nrc3557

Ludwig C, Viant MR (2010) Two-dimensional J-resolved NMR spectroscopy: review of a key methodology in the metabolomics toolbox. Phytochem Anal. 21:22-32. doi: 10.1002/pca.1186.

Lushchak VI (2012) Glutathione homeostasis and functions: potential targets for medical interventions. J Amino Acids 2012:736837. doi: 10.1155/2012/736837

Matulewicz Ł, Cichoń A, Sokół M, Przybyszewski W, Głowala-Kosińska M, Gibas M (2013) High resolution proton nuclear magnetic resonance (1H NMR) spectroscopy of surviving C6 glioma cells after X-ray irradiation. Folia Neuropathol 51:33-43.

Milkevitch M, Shim H, Pilatus U, Pickup S, Wehrle JP, Samid D, Poptani H, Glickson JD, Delikatny EJ (2005) Increases in NMR-visible lipid and glycerophosphocholine during phenylbutyrate-induced apoptosis in human prostate cancer cells. Biochim Biophys Acta 1734:1-12. doi: 10.1016/j.bbalip.2005.01.008

Monceau V, Meziani L, Strup-Perrot C, Morel E, Schmidt M, Haagen J, Escoubet B, Dörr W, Vozenin MC (2013) Enhanced sensitivity to low dose irradiation of ApoE-/- mice mediated by early pro-inflammatory profile and delayed activation of the TGFβ1 cascade involved in fibrogenesis. PLoS One 8:e57052. doi: 10.1371/journal.pone.0057052

Morán J, Hernández-Pech X, Merchant-Larios H, Pasantes-Morales H (2000) Release of taurine in apoptotic cerebellar granule neurons in culture. Pflugers Arch 439:271-7.

Nakae I, Mitsunami K, Omura T, Yabe T, Tsutamoto T, Matsuo S, Takahashi M, Morikawa S, Inubushi T, Nakamura Y et al (2003) Proton magnetic resonance spectroscopy can detect creatine depletion associated with the progression of heart failure in cardiomyopathy. J Am Coll Cardiol 42:1587-93.

Palma A, Grande S, Luciani AM, Mlynárik V, Guidoni L, Viti V, Rosi A (2016) Metabolic Study of Breast MCF-7 Tumor Spheroids after Gamma Irradiation by (1)H NMR Spectroscopy and Microimaging. Front Oncol 6:105. doi: 10.3389/fonc.2016.00105

Pan X, Wilson M, Mirbahai L, McConville C, Arvanitis TN, Griffin JL, Kauppinen RA, Peet AC (2011) In vitro metabonomic study detects increases in UDP-GlcNAc and UDP-GalNAc, as early phase markers of cisplatin treatment response in brain tumor cells. J Proteome Res. 10:3493-500. doi: 10.1021/pr200114v

Pathak CM, Avti PK, Kumar S, Khanduja KL, Sharma SC (2007) Whole body exposure to low-dose gamma radiation promotes kidney antioxidant status in Balb/c mice. J Radiat Res 48:113-20.

Perry DK, Carton J, Shah AK, Meredith F, Uhlinger DJ, Hannun YA (2000) Serine palmitoyltransferase regulates de novo ceramide generation during etoposide-induced apoptosis. J BiolChem 275:9078-84.

Prosnitz RG, Hubbs JL, Evans ES, Zhou SM, Yu X, Blazing MA, Hollis DR, Tisch A, Wong TZ, Borges-Neto S, Hardenbergh PH, Marks LB (2007) Prospective Assessment of Radiotherapy associated Cardiac Toxicity in Breast Cancer Patients: Analysis of Data 3 to 6 Years After Treatment. Cancer 110:1840-1850.

Rainaldi G, Romano R, Indovina P, Ferrante A, Motta A, Indovina PL, Santinia MT (2008) Metabolomics using 1H-NMR of apoptosis and Necrosis in HL60 leukemia cells: differences between the two types of cell death and independence from the stimulus of apoptosis used. Radiat Res 169:170-80doi: 10.1667/RR0958.1

Rhee EP, Gerszten RE (2012) Metabolomics and cardiovascular biomarker discovery. Clin Chem 58:139-47. doi: 10.1373/clinchem.2011.169573

Ripps H, Shen W (2012) Review: taurine: a "very essential" amino acid. Mol Vis 18:2673-86.

Santini MT, Romano R, Rainaldi G, Ferrante A, Indovina P, Motta A, Indovina PL (2006a) 1H-NMR evidence for a different response to the same dose (2 Gy) of ionizing radiation of MG-63 human osteosarcoma cells and three-dimensional spheroids. Anticancer Res 26:267-81.

Santini MT, Romano R, Rainaldi G, Indovina P, Ferrante A, Motta A, Indovina PL (2006b) Temporal dynamics of 1H-NMR-visible metabolites during radiation-induced apoptosis in MG-63 human osteosarcoma spheroids. Radiat Res 166:734-45. doi: 10.1667/RR0635.1

Schaffer SW, Jong CJ, Ramila KC, Azuma J (2010) Physiological roles of taurine in heart and muscle. J Biomed Sci 17 Suppl 1:S2. doi: 10.1186/1423-0127-17-S1-S2

Schnackenberg LK, Pence L, Vijay V, Moland CL, George N, Cao Z, Yu LR, Fuscoe JC, Beger RD, Desai VG (2016) Early metabolomics changes in heart and plasma during chronic doxorubicin treatment in B6C3F1 mice. J Appl Toxicol 36:1486-95. doi: 10.1002/jat.3307

Schwenen M, Altman KI, Schröder W (1989) Radiation-induced increase in the release of amino acids by isolated, perfused skeletal muscle. Int J Radiat Biol 55:257-69.

Shimzu T, Iwanaga M, Yasunaga A, Urata Y, Goto S, Shibata S, Kondo T (1998) Protective role of glutathione synthesis on radiation-induced DNA damage in rabbit brain. Cell Mol Neurobiol 18:299-310.

Shimizu Y, Kodama K, Nishi N, Kasagi F, Suyama A, Soda M, Grant EJ, Sugiyama H, Sakata R, Moriwaki H, Hayashi M, Konda M, Shore RE (2010) Radiation exposure and circulatory disease risk: Hiroshima and Nagasaki atomic bomb survivor data, 1950-2003. BMJ 340:b5349. doi: 10.1136/bmj.b5349

Sokół M, Przybyszewski WM, Matlas B (2004) Investigation of metabolic changes in irradiated rat brain tissue by means of 1H NMR in vitro relaxation study. Solid State Nucl Magn Reson 25:53-60. doi: 10.1016/j.ssnmr.2003.03.013

Thurston JH, Hauhart RE, Naccarato EF (1981) Taurine: possible role in osmotic regulation of mammalian heart. Science 214:1373-4.

Tom A, Nair KS (2006) Assessment of branched-chain amino Acid status and potential for biomarkers. J Nutr 136(1 Suppl):324S-30S

Vasconcelos-Dos-Santos A, Oliveira IA, Lucena MC, Mantuano NR, Whelan SA, Dias WB, Todeschini AR (2015) Biosynthetic Machinery Involved in Aberrant Glycosylation: Promising Targets for Developing of Drugs Against Cancer. Front Oncol 5:138. doi: 10.3389/fonc.2015.00138

Wells L, Vosseller K, Hart GW (2003) A role for N-acetylglucosamine as a nutrient sensor and mediator of insulin resistance. Cell Mol Life Sci 60:222-8.

Xiao X, Hu M, Liu M, Hu JZ (2016) 1H NMR Metabolomics Study of Spleen from C57BL/6 Mice Exposed to Gamma Radiation. Metabolomics (Los Angel) 6:1-11. doi: 10.4172/2153-0769.1000165

Zervou S, Whittington HJ, Russell AJ, Lygate CA (2016) Augmentation of Creatine in the Heart. Mini Rev Med Chem 16:19-28.

Published
2018-07-08
Section
Articles