Skip to main content
Log in

Curative role of pantothenic acid in brain damage of gamma irradiated rats

  • Original Article
  • Published:
Indian Journal of Clinical Biochemistry Aims and scope Submit manuscript

Abstract

Radiation induced brain damage is associated with impairment of mitochondrial functions, variations in the level of neurotransmitters, and oxidative stress. Mitochondrial function is closely linked to the level of neurotransmitters since the precursors are supplied by the Kreb’s cycle intermediates. The objective of this study was to evaluate the influence of pantothenic acid, an essential component in the synthesis of Coenzyme A (CoA), on the activity of the Krebs cycle enzymes, isocitrate dehydrogenase (IDH), α-ketoglutarate dehydrogenase (α-KGDH), and succinate dehydrogenase (SDH); the level of aspartic, glutamic and GABA; the activity of transaminases, and oxidative stress, in the cerebrum of γ-irradiated rats. Pantothenic acid (26 mg/Kg) was orally administered to the rats, 2 h after irradiation and during the following 5 days. Animals were sacrificed the 7th day post-irradiation. The exposure of male albino rats to γ-rays (5 Gy) has triggered oxidative stress notified by a significant elevation in the level of malondialdehyde (MDA), an end product of lipid peroxidation, associated to a significant decrease in the content of phospholipids, and the antioxidant compound glutathione (GSH). The activity of IDH, α-KGDH, and SDH, has significantly decreased, while the level of aspartic, glutamic and GABA has significantly increased. In parallel to these changes, the activity of alanine and aspartate transaminase has significantly increased, compared to their values in the control rats. Pantothenic acid treatment, has significantly attenuated oxidative stress; enhanced the activity of IDH, α-KGDH, and SDH; minimized the increase in the level of amino acids and the activity of transaminases, compared to their values in the cerebrum of irradiated rats. In conclusion, pantothenic acid could improve the level of neurotransmitters amino acids, which depends on the enzymatic activities of Krebs cycle and linked to oxidative stress.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Ward JF. The yield of DNA double-strand breaks produced intracellularly by ionizing radiation: a review. Int J Radiat Biol. 1990;57(6):1141–50.

    Article  PubMed  CAS  Google Scholar 

  2. Panganiban RM, Snow AL, Day RM. Mechanisms of radiation toxicity in transformed and non-transformed cells. Int J Mol Sci. 2013;14(8):15931–58.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Saada HN, Said UZ, Mahdy EM, Elmezayen HE, Shedid SM. Fish oil omega-3 fatty acids reduce the severity of radiation-induced oxidative stress in the rat brain. Int J Radiat Biol. 2014;90(12):1179–83.

    Article  PubMed  CAS  Google Scholar 

  4. Zuin A, Gabrielli N, Calvo IA, Garcia-Santamarina S, Hoe KL, Kim DU, et al. Mitochondrial dysfunction increases oxidative stress and decreases chronological life span in fission yeast. PLoS ONE. 2008;3:2842.

    Article  CAS  Google Scholar 

  5. Lu SC. Regulation of glutathione synthesis. Mol Asp Med. 2009;30(1–2):42–59.

    Article  CAS  Google Scholar 

  6. Schulz JB, Lindenau J, Seyfried J, Dichgans J. Glutathione, oxidative stress and neurodegeneration. Eur J Biochem. 2000;267:4904–11.

    Article  PubMed  CAS  Google Scholar 

  7. Tiwari V, Ambadipudi S, Patel AB. Glutamatergic and GABAergic TCA cycle and neurotransmitter cycling fluxes in different regions of mouse brain. J Cereb Blood Flow Metab. 2013;33(10):1523–31.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Thompson CB. Metabolic enzymes as oncogenes or tumor suppressors. N Engl J Med. 2009;360(8):813–5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Maus A, Peters GJ. Glutamate and α-ketoglutarate: key players in glioma metabolism. Amino Acids. 2017;49(1):21–32.

    Article  PubMed  CAS  Google Scholar 

  10. Zhao S, Lin Y, Xu W, Jiang W, Zha Z, Wang P, et al. Glioma-derived mutations in IDH1 dominantly inhibit IDH1 catalytic activity and induce HIF-1α. Science. 2009;324(5924):261–5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Gabriel JL, Zervos PR, Plaut GW. Activity of purified NAD-specific isocitrate dehydrogenase at modulator and substrate concentrations approximating conditions in mitochondria. Metabolism. 1986;35(7):661–7.

    Article  PubMed  CAS  Google Scholar 

  12. Fujii T, Khawaja MR, DiNardo CD, Atkins JT, Janku F. Targeting isocitrate dehydrogenase (IDH) in cancer. Discov Med. 2016;21(117):373–80.

    PubMed  Google Scholar 

  13. Viswanath P, Chaumeil MM, Ronen SM. Molecular imaging of metabolic reprograming in mutant IDH cells. Front Oncol. 2016;6:60.

    Article  PubMed  PubMed Central  Google Scholar 

  14. McLain AL, Cormier PJ, Kinter M, Szweda LI. Glutathionylation of α-ketoglutarate dehydrogenase: the chemical nature and relative susceptibility of the cofactor lipoic acid to modification. Free Radic Biol Med. 2013;61:161–9.

    Article  PubMed  CAS  Google Scholar 

  15. Tretter L, Adam-Vizi V. Alpha-ketoglutarate dehydrogenase: a target and generator of oxidative stress. Philos Trans R Soc Lond B Biol Sci. 2005;360(1464):2335–45.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Jeitner TM, Xu H, Gibson GE. Inhibition of the alpha-ketoglutarate dehydrogenase complex by the myeloperoxidase products, hypochlorous acid and mono-N-chloramine. J Neurochem. 2005;92:302–10.

    Article  PubMed  CAS  Google Scholar 

  17. Gibson GE, Blass JP, Beal MF, Bunik V. The alpha-ketoglutarate dehydrogenase complex: a mediator between mitochondria and oxidative stress in neurodegeneration. Mol Neurobiol. 2005;31(1–3):43–63.

    Article  PubMed  CAS  Google Scholar 

  18. Ko LW, Sheu KF, Thaler HT, Markesbery WR, Blass JP. Selective loss of KGDHC-enriched neurons in Alzheimer temporal cortex: Does mitochondrial variation contribute to selective vulnerability? J Mol Neurosci. 2001;17:361–9.

    Article  PubMed  CAS  Google Scholar 

  19. Gibson GE, Kingsbury AE, Xu H, Lindsay JG, Daniel S, Foster OJ, et al. Deficits in a Krebs acid cycle enzyme in brains from patients with Parkinson’s disease. Neurochem Int. 2003;43:129–35.

    Article  PubMed  CAS  Google Scholar 

  20. Oyedotun KS, Lemire BD. The quaternary structure of the Saccharomyces cerevisiae succinate dehydrogenase. Homology modeling, cofactor docking, and molecular dynamics simulation studies. J Biol Chem. 2004;279(10):9424–31.

    Article  PubMed  CAS  Google Scholar 

  21. Huang S, Milla AH. Succinate dehydrogenase: the complex roles of a simple enzyme. Curr Opin Plant Biol. 2013;16(3):344–9.

    Article  PubMed  CAS  Google Scholar 

  22. Skillings EA, Morton AJ. Delayed onset and reduced cognitive deficits through pre-conditioning with 3-nitropropionic acid is dependent on sex and CAG repeat length in the r6/2 mouse model of Huntington’s disease. J Huntingt Dis. 2016;5(1):19–32.

    Article  CAS  Google Scholar 

  23. Ayano G. Common neurotransmitters: criteria for neurotransmitters, key locations, classifications and functions. APN. 2016;1(1):1–5.

    Google Scholar 

  24. Kendra C. What is a neurotransmitter?. In: The everything psychology book. 2nd ed. USA: Adams Media Corporation (Kindle Edition); 2014.

  25. Sapolsky R. Biology and human behavior: the neurological origins of individuality. In: Guide book. 2nd ed. Chantilly: The Teaching Company; 2005. pp 13–14.

  26. Park CH, Choi SH, Piao Y, Kim S, Lee YJ, Kim HS, et al. Glutamate and aspartate impair memory retention and damage hypothalamic neurons in adult mice. Toxicol Lett. 2000;115:117–25.

    Article  PubMed  CAS  Google Scholar 

  27. Dong XX, Wang Y, Qin ZH. Molecular mechanisms of excitotoxicity and their relevance to pathogenesis of neurodegenerative disease. Acta Pharmacol Sin. 2009;30:379–87.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Hynd M, Scott HL, Dodd PR. Glutamate-mediated excitotoxicity and neurodegeneration in Alzheimer’s disease. Neurochem Int. 2004;45(5):583–95.

    Article  PubMed  CAS  Google Scholar 

  29. Blandini F, Porter RH, Greenamyre JT. Glutamate and Parkinson’s disease. Mol Neurobiol. 1996;12:73–94.

    Article  PubMed  CAS  Google Scholar 

  30. Sanacora G, Gueorguieva R, Epperson CN, Wu YT, Appel M, Rothman DL. Subtype-specific alterations of gamma-aminobutyric acid and glutamate in patients with major depression. Arch Gen Psychiatry. 2004;61:705–13.

    Article  PubMed  CAS  Google Scholar 

  31. Chen PE, Geballe MT, Stansfeld PJ, Johnston AR, Yuan H, Jacob AL, et al. Structural features of the glutamate binding site in recombinant NR1/NR2A N-methyl-d-aspartate receptors determined by site-directed mutagenesis and molecular modeling. Mol Pharmacol. 2005;67(5):1470.

    Article  PubMed  CAS  Google Scholar 

  32. D’Aniello S, Somorjai I, Garcia-Fernàndez J, Topo E, D’Aniello A. d-aspartic acid is a novel endogenous neurotransmitter. FASEB J. 2011;25(3):1014–27.

    Article  PubMed  CAS  Google Scholar 

  33. Errico F, Rossi S, Napolitano F, Catuogno V, Topo E, Fisone G, et al. d-aspartate prevents corticostriatal long-term depression and attenuates schizophrenia-like symptoms induced by amphetamine and MK-801. J Neurosci. 2008;28:10404–14.

    Article  PubMed  CAS  Google Scholar 

  34. Topo E, Soricelli A, Di Maio A, D’Aniello E, Di Fiore MM, D’Aniello A. Evidence for the involvement of d-aspartic acid in learning and memory of rat. Amino Acids. 2010;38:1561–9.

    Article  PubMed  CAS  Google Scholar 

  35. Kim PM, Duan X, Huang AS, Liu CY, Ming G, Song H, et al. Aspartate racemase, generating neuronal d-aspartate, regulates adult neurogenesis. Proc Natl Acad Sci. 2010;107(7):3175–9.

    Article  PubMed  Google Scholar 

  36. Uchida Y, Ito K, Ohtsuki S, Kubo Y, Suzuki T, Terasaki T. Major involvement of Na + -dependent multivitamin transporter (SLC5A6/SMVT) in uptake of biotin and pantothenic acid by human brain capillary endothelial cells. J Neurochem. 2015;134:97–112.

    Article  PubMed  CAS  Google Scholar 

  37. Kennedy DO. B vitamins and the brain: mechanisms, dose and efficacy—a review. Nutrients. 2016;8(2):68.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Rucker RB, Bauerly K. Pantothenic acid. In: Zempleni J, Suttie JW, Gregory III JF, Stover PJ, editors. Handbook of Vitamins. 5th ed. Boca Raton: CRC Press; 2013.

    Google Scholar 

  39. Gominak SC. Vitamin D deficiency changes the intestinal microbiome reducing B vitamin production in the gut. The resulting lack of pantothenic acid adversely affects the immune system, producing a “pro-inflammatory” state associated with atherosclerosis and autoimmunity. Med Hypotheses. 2016;94:103–7.

    Article  PubMed  CAS  Google Scholar 

  40. Otten JJ, Hellwig JP, Meyers LD. Dietary reference intakes: the essential guide to nutrient requirements. Washington: The National Academies Press; 2008.

    Google Scholar 

  41. Beers MH, Berkow R. Biology of the immune system. In: Beers MH, Berkow R, editors. The Merck manual of diagnosis and therapy. 17th ed. Washington: Whitehouse; 1999.

  42. Vorobev VV, Moiseenok AG, Khomich TI. Characteristics of pantothenic acid transport in membrane vesicles of the brush border of small intestine epithelial cells in rats. Ross Fiziol Zh Im IM Sechenova. 1998;84(8):814–20.

    CAS  Google Scholar 

  43. Schroeder HA. Losses of vitamins and trace minerals resulting from processing and preservation of foods. Am J Clin Nutr. 1971;24:562–73.

    Article  PubMed  CAS  Google Scholar 

  44. Said UZ, Saada HN, Abd-Alla MS, Elsayed ME, Amin AM. Hesperidin attenuates brain biochemical changes of irradiated rats. Int J Radiat Biol. 2012;88:613–8.

    Article  PubMed  CAS  Google Scholar 

  45. Slyshenkov VS, Omelyanchik SN, Moiseenok AG, Trebukhina RV, Wojtczak L. Pantothenol protects rats against some deleterious effects of gamma radiation. Free Radic Biol Med. 1998;24(6):894–9.

    Article  PubMed  CAS  Google Scholar 

  46. Yoshioka T, Kawada K, Shimada T, Mori M. Lipid peroxidation in maternal and cord blood and protective mechanism against activated oxygen toxicity in the blood. Am J Obstet Gynecol. 1979;135:372–6.

    Article  PubMed  CAS  Google Scholar 

  47. Beutler E, Duron O, Kelly BM. Improved method for determination of blood glutathione. J Lab Clin Med. 1963;61:882–8.

    PubMed  CAS  Google Scholar 

  48. Connerty HV, Briggs AR, Eaton EH. Simplified determination of the lipid components of blood serum. Clin Chem Acta. 7(1):37–53. Determination of serum, phospholipids, lipid phosphorous. In: Varley, H, editor. Practical clinical biochemistry. 4th ed. India: CBS Publishers; 1961. pp. 319–320.

  49. Rickwood D, Wilson MT, Darley-Usmar VM. Isolation and characteristic of intact mitochondria. In: Darley-Usmar VM, Rickwood D, Wilson MT, editors. Mitochondria: a practical approach. Washington: I. R. L. Press; 1978. p. 4–5.

    Google Scholar 

  50. Kim HJ, Winge DR. Emerging concepts in the flavinylation of succinate dehydrogenase. Biochim Biophys Acta. 2013;1827:627–36.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Hsu JM, Anthony WL, Rider AA. Free amino acids in plasma and tissue of rats born to underfed dams. Proc Soc Exp Biol Med. 1975;148:1087.

    Article  PubMed  CAS  Google Scholar 

  52. Henry RJ, Chiamori N, Golub OJ, Berkman S. Revised spectrophotometric methods for the determination of glutamic-oxalacetic transaminase, glutamic-pyruvic transaminase, and lactic acid dehydrogenase. Am J Clin Path. 1960;34:381–98.

    Article  PubMed  CAS  Google Scholar 

  53. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem. 1951;193(1):265–75.

    PubMed  CAS  Google Scholar 

  54. Sherki Y, Melamed E, Offen D. Oxidative stress induced neurodegenerative diseases: the need for antioxidants that penetrate the blood brain barrier. Neuropharmacology. 2001;40:959.

    Article  Google Scholar 

  55. Spitz DR, Azzam EI, Li JJ, Gius D. Metabolic oxidation/reduction reactions and cellular responses to ionizing radiation: a unifying concept in stress response biology. Cancer Metastasis Rev. 2004;23:311–22.

    Article  PubMed  CAS  Google Scholar 

  56. Nordberg J, Arner ESJ. Reactive oxygen species, antioxidants, and the mammalian thioredoxin system. Free Radic Biol Med. 2001;31:1287–312.

    Article  PubMed  CAS  Google Scholar 

  57. Mailloux RJ, Beriault R, Lemire J, Singh R, Chenier DR, Hamel RD, et al. The Krebs acid cycle, an ancient metabolic network with a novel twist. PLoS ONE. 2007;2:e690.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Scornavacca G, Gesuete R, Orsini F, Pastorelli R, Fanelli R, de Simoni MG, et al. Proteomic analysis of mouse brain cortex identifies metabolic down-regulation as a general feature of ischemic pre-conditioning. J Neurochem. 2012;122(6):1219–29.

    Article  PubMed  CAS  Google Scholar 

  59. Mailloux RJ, Singh R, Brewer G, Auger C, Lemire J, Appanna VD. α-ketoglutarate dehydrogenase and glutamate dehydrogenase work in tandem to modulate the antioxidant α-ketoglutarate during oxidative stress in pseudomonas fluorescens. J Bacteriol. 2009;191(12):3804–10.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Hutchinson P, O’Connell M, Al-Rawi P, Kett-White C, Gupta A, Maskell L, et al. Increases in GABA concentrations during cerebral ischaemia: a microdialysis study of extracellular amino acids. J Neurol Neurosurg Psychiatry. 2002;72(1):99–105.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Healy D. Serotonin and depression. BMJ. 2015;350:1771.

    Article  Google Scholar 

  62. Trofimova I, Robbins TW. Temperament and arousal systems: a new synthesis of differential psychology and functional neurochemistry. Neurosci Biobehav Rev. 2016;64:382–402.

    Article  PubMed  CAS  Google Scholar 

  63. Trofimova IN. The interlocking between functional aspects of activities and a neurochemical model of adult temperament. In: Arnold MC, editor. Temperaments: individual differences, social and environmental influences and impact on quality of life. New York: Nova Science Publishers, Inc; 2016. p. 77–147.

    Google Scholar 

  64. Scientific Committee on Food. Opinion of the Scientific Committee on Food on the tolerable upper intake level of pantothenic acid. Report of the European Commission: health and consumer protection directorate-general 2002SCF/CS/NUT/UPPLEV/61 Final:1-6. 2011. http://ec.europa.eu/food/fs/sc/scf/out80k_en.pdf.

  65. Shibata K, Takahashi C, Fukuwatari T, Sasaki R. Effects of excess pantothenic acid administration on the other water-soluble vitamin metabolisms in rats. J Nutr Sci Vitaminol. 2005;51:385–91.

    Article  PubMed  CAS  Google Scholar 

  66. Moorthy PN, Hayon E. One-electron redox reactions of water-soluble vitamins. IV. Thiamin (vitamin B1), biotin, and pantothenic acid. J Org Chem. 1977;42(5):879–85.

    Article  PubMed  CAS  Google Scholar 

  67. Akram E, Pejman M, Masoud ET, Ali HR, Shahabaldin S. Hepatoprotective effects of pantothenic acid on carbon tetrachloride-induced toxicity in rats. EXCLI J. 2012;11:748–59.

    Google Scholar 

  68. Demirci B, Demir O, Dost T, Birincioglu M. Protective effect of vitamin B5 (dexpanthenol) on cardiovascular damage induced by streptozocin in rats. Bratisl Lek Listy. 2014;115(4):190–6.

    PubMed  CAS  Google Scholar 

  69. Wang B, Zhang X, Yue B, Ge W, Zhang M, Ma C, et al. Effects of pantothenic acid on growth performance, slaughter performance, lipid metabolism, and antioxidant function of Wulong geese aged one to four weeks. Anim Nutr. 2016;2(4):312–7.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Slyshenkov VS, Dymkowska D, Wojtczak L. Pantothenic acid and pantothenol increase biosynthesis of glutathione by boosting cell energetics. FEBS Lett. 2004;569(1–3):169–72.

    Article  PubMed  CAS  Google Scholar 

  71. Saada HN, Rezk RG, Eltahawy NA. Lycopene protects the structure of the small intestine against gamma-radiation-induced oxidative stress. Phytother Res. 2010;24(2):204–8.

    Article  Google Scholar 

  72. Daugherty M, Polanuyer B, Farrell M, Scholle M, Lykidis A, de Crécy-Lagard V, et al. Complete reconstitution of the human coenzyme a biosynthetic pathway via comparative genomics. J Biol Chem. 2002;277:21431–9.

    Article  PubMed  CAS  Google Scholar 

  73. Voet D, Voet JG, Pratt CW. Fundamentals of biochemistry: life at the molecular level. 2nd ed. Hoboken: Wiley; 2006.

    Google Scholar 

  74. Gropper SS, Smith JL, Groff JL. Advanced nutrition and human metabolism. Belmont: Wadsworth, Cengage Learning; 2009.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Shedid SM.

Ethics declarations

Conflict of interest

The authors report no conflict of interest.

Ethical approval

All the experimental procedures were carried out according to the principles and guidelines of the Ethics Committee of the National Research Centre conformed to “Guide for the care and use of Laboratory Animals” for the use and welfare of experimental animals, published by the US National Institutes of Health (NIH publication No. 85–23, 1996).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

SM, S., HN, S., NA, E. et al. Curative role of pantothenic acid in brain damage of gamma irradiated rats. Ind J Clin Biochem 33, 314–321 (2018). https://doi.org/10.1007/s12291-017-0683-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12291-017-0683-0

Keywords

Navigation