Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter September 21, 2011

Alterations in anti-oxidative defence enzymes in erythrocytes from sporadic amyotrophic lateral sclerosis (SALS) and familial ALS patients

  • Aleksandra Nikolić-Kokić , Zorica Stević , Duško Blagojević , Biljana Davidović , David R. Jones and Mihajlo B. Spasić

Abstract

Background: Overproduction of nitric oxide (NO) and hydrogen peroxide (H2O2) may be an important factor in the pathogenesis of amyotrophic lateral sclerosis (ALS). Owing to their ability to permeate through biological membranes, excess NO and H2O2 may be present in the media surrounding motor neurones. Anti-oxidative defence enzymes (ADEs) in erythrocytes are capable of detoxifying reactive oxygen species (produced endogenously or exogenously), but may also be structurally modified and inactivated by reactive oxygen and nitrogen species. Both balanced and coordinated ADE activities are of utmost importance for their correct physiological function.

Methods: We determined activity of the following ADEs: copper-zinc superoxide dismutase (CuZn SOD), catalase (CAT), glutathione peroxidase (GSH-Px) and glutathione reductase (GR) in erythrocytes from sporadic ALS patients [SALS (−/+)], familial ALS patients with the Leu144Phe mutation in the SOD1 gene [FALS (+/+)], asymptomatic carriers with the Leu144Phe mutation in the SOD1 gene (+/−), and control subjects (−/−). We also examined the in vitro effect of diethyldithiocarbamate (DDC) on CuZn SOD activity in erythrocytes from FALS patients, SALS patients and control subjects.

Results: The influence of the Leu144Phe mutation and/or disease was apparent for ADE activities measured in all three patient groups. The SOD1 gene mutation decreased CuZn SOD and GSH-Px activity (two-way ANOVA, significant mutation effect). We noted that the disease also contributed to decreased CuZn SOD activity in SALS patients in comparison with the control group (two-way ANOVA, mutation and disease effect). The disease also influenced CAT and GR activity. CAT activity was decreased in both SALS and FALS patients. In all three patient groups, GR activity was higher than in the control group. Finally, DDC inhibited CuZn SOD activity in erythrocytes from control subjects, FALS (Leu144Phe) patients and SALS patients; however, its effect was more pronounced and significant in FALS patients.

Conclusions: Changes in erythrocyte ADE activities suggest that oxidative stress, involved in the motor neurone pathogenesis of SALS and FALS, also has systemic effects. Differences in ADE systems between the study groups revealed the presence of different types of oxidative pressure, indicating the potential additional benefit of individually designed anti-oxidant cocktail therapies.


Corresponding author: Aleksandra Nikolić-Kokić, Department of Physiology, Institute for Biological Research “Siniša Stanković”, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia and Montenegro Phone: +381-11-2078348, Fax: +381-11-761422,

References

1. Patterson D, Warner HR, Fox LM, Rahmani Z. Superoxide dismutase, oxygen radical metabolism, and amyotrophic lateral sclerosis. Mol Genet Med 1994; 4:79–118.10.1016/B978-0-12-462004-9.50007-4Search in Google Scholar

2. Tohgi H, Abe T, Yamazaki K, Murata T, Ishizaki E, Isobe C. Increase in oxidized NO products and reduction in oxidized glutathione in cerebrospinal fluid from patients with sporadic form of amyotrophic lateral sclerosis. Neurosci Lett 1999; 260:204–6.10.1016/S0304-3940(98)00986-0Search in Google Scholar

3. Nikolić Kokić A, Stević Z, Stojanović S, Blagojević PD, Jones DR, Pavlović S, et al. Biotransformation of nitric oxide in the cerebrospinal fluid of amyotrophic lateral sclerosis patients. Redox Rep 2005; 10:265–70.10.1179/135100005X70242Search in Google Scholar

4. Przedborski S, Donaldson DM, Murphy PL, Hirsch O, Lange D, Naini AB, et al. Blood superoxide dismutase, catalase and glutathione peroxidase activities in familial and sporadic amyotrophic lateral sclerosis. Neurodegeneration 1996; 5:57–64.10.1006/neur.1996.0008Search in Google Scholar

5. Apostolski S, Marinkovic Z, Nikolic A, Blagojevic D, Spasic MB, Michelson AM. Glutathione peroxidase in amyotrophic lateral sclerosis: the effects of selenium supplementation. J Environ Pathol Toxicol Oncol 1998; 17:325–9.Search in Google Scholar

6. Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, Hentati A, et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 1993; 362:59–62.10.1038/362059a0Search in Google Scholar

7. Bonnefont-Rousselot D, Lacomblez L, Jaudon M, Lepage S, Salachas F, Bensimon G, et al. Blood oxidative stress in amyotrophic lateral sclerosis. J Neurol Sci 2000; 178:57–62.10.1016/S0022-510X(00)00365-8Search in Google Scholar

8. Beal MF, Ferrante RJ, Browne SE, Matthews RT, Kowall NW, Brown RH Jr. Increased 3-nitrotyrosine in both sporadic and familial amyotrophic lateral sclerosis. Ann Neurol 1997; 42:644–54.10.1002/ana.410420416Search in Google Scholar

9. Bruijn LI, Beal MF, Becher MW, Schulz JB, Wong PC, Price DL, et al. Elevated free nitrotyrosine levels, but not protein-bound nitrotyrosine or hydroxyl radicals, throughout amyotrophic lateral sclerosis (ALS)-like disease implicate tyrosine nitration as an aberrant in vivo property of one familial ALS-linked superoxide dismutase 1 mutant. Proc Natl Acad Sci USA 1997; 94:7606–11.10.1073/pnas.94.14.7606Search in Google Scholar

10. Andrus PK, Fleck TJ, Gurney ME, Hall ED. Protein oxidative damage in a transgenic mouse model of familial amyotrophic lateral sclerosis. J Neurochem 1998; 71:2041–8.10.1046/j.1471-4159.1998.71052041.xSearch in Google Scholar

11. Hall ED, Andrus PK, Oostveen JA, Fleck TJ, Gurney ME. Relationship of oxygen radical-induced lipid peroxidative damage to disease onset and progression in a transgenic model of familial ALS. J Neurosci Res 1998; 53:66–77.10.1002/(SICI)1097-4547(19980701)53:1<66::AID-JNR7>3.0.CO;2-HSearch in Google Scholar

12. Liochev SI, Fridovich I. Copper- and zinc-containing superoxide dismutase can act as a superoxide reductase and a superoxide oxidase. J Biol Chem 2000; 275:38482–5.10.1074/jbc.M007891200Search in Google Scholar

13. Durham HD, Roy J, Dong L, Figlewich DA. Aggregation of mutant Cu/Zn superoxide dismutase proteins in a culture model of ALS. J Neuropathol Exp Neurol 1997; 56:523–30.10.1097/00005072-199705000-00008Search in Google Scholar

14. Estevez AG, Crow JP, Sampson JB, Reiter C, Zhuang Y, Richardson GJ, et al. Induction of nitric oxide-dependent apoptosis in motor neurons by zinc-deficient superoxide dismutase. Science 1999; 286:2498–500.10.1126/science.286.5449.2498Search in Google Scholar

15. Wiedau-Pazos M, Goto JJ, Rabizadeh S, Gralla EB, Roe JA, Lee MK, et al. Altered reactivity of superoxide dismutase in familial amyotrophic lateral sclerosis. Science 1996; 271:515–8.10.1126/science.271.5248.515Search in Google Scholar

16. Simpson EP, Yen AA, Appel SH. Oxidative stress: a common denominator in the pathogenesis of amyotrophic lateral sclerosis. Curr Opin Rheumatol 2003; 15:730–6.10.1097/00002281-200311000-00008Search in Google Scholar

17. Chance B, Sies H, Boveris A. Hydroperoxide metabolism in mammalian organs. Physiol Rev 1979; 59:527–605.10.1152/physrev.1979.59.3.527Search in Google Scholar

18. Gul M, Kutay FZ, Temocin S, Hanninen O. Cellular and clinical implications of glutathione. Indian J Exp Biol 2000; 38:625–34.Search in Google Scholar

19. Andersen HR, Nielsen JB, Nielsen F, Grandjean P. Antioxidative enzyme activity in human erythrocytes. Clin Chem 1997; 43:562–8.10.1093/clinchem/43.4.562Search in Google Scholar

20. Sinet PM, Lavelle F, Michelson AM, Jerome H. Superoxide dismutase activities of blood platelets in Trisomy 21. Biochem Biophys Res Commun 1975; 67:904–9.10.1016/0006-291X(75)90762-7Search in Google Scholar

21. Sinet PM, Michelson AM, Bazin A, Lejeune J, Jerome H. Increase in glutathione peroxidase activity in erythrocytes from Trisomy 21 subjects. Biochem Biophys Res Commun 1975; 67:910–5.10.1016/0006-291X(75)90763-9Search in Google Scholar

22. Pastor MC, Sierra C, Dolade M, Navarro E, Brandi N, Cabre E, et al. Antioxidant enzymes and fatty acid status in erythrocytes of Down syndrome patients. Clin Chem 1998; 44:924–9.10.1093/clinchem/44.5.924Search in Google Scholar

23. Ferri A, Nencini M, Casciati A, Cozzolino M, Angelini DF, Longone P, et al. Cell death in amyotrophic lateral sclerosis: interplay between neuronal and glial cells. FASEB J 2004; 18:1261–3.10.1096/fj.03-1199fjeSearch in Google Scholar

24. Brooks BR. El escorial World Federation of Neurology criteria for the diagnosis of amyotrophic lateral sclerosis. J Neurol Sci 1994; 124(Suppl 1):96–107.10.1016/0022-510X(94)90191-0Search in Google Scholar

25. Norris FH Jr, Calanchini PR, Fallat RJ, Panchari S, Jewett B. The administration of guanidine in amyotrophic lateral sclerosis. Neurology 1974; 24:721–8.10.1212/WNL.24.8.721Search in Google Scholar

26. Tsuchihashi M. Zur Kenntnis der Blutkatalase. Biochem Z 1923; 140:65–74.Search in Google Scholar

27. McCord JM, Fridovich I. Superoxide dismutase. An enzymic function for erythrocuprein (hemocuprein). J Biol Chem 1969; 244:6049–55.Search in Google Scholar

28. Beutler E. Catalase: a manual of biochemical methods. In: Beutler E, editor. Red cell metabolism. New York: Grune and Stratton, 1982:105–6.Search in Google Scholar

29. Paglia DE, Valentine WN. Studies on the quantitative and qualitative characterization of erythrocyte glutathione peroxidase. J Lab Clin Med 1967; 70:74–7.Search in Google Scholar

30. Glatzle D, Vuilleumier JP, Weber F, Decker K. Glutathione reductase test with whole blood: a convenient procedure for the assessment of the riboflavin status in humans. Experientia 1974; 30:665–8.10.1007/BF01921531Search in Google Scholar

31. Drabkin D, Austin H. Spectrophotometric studies: preparations from washed blood cells. J Biol Chem 1935; 112:51–5.10.1016/S0021-9258(18)74965-XSearch in Google Scholar

32. Hinkle ED, Wiersma W, Jurs GS. Applied statistics for behavioral sciences, 3rd ed. Boston, MA: Houghton Mifflin Company, 1994.Search in Google Scholar

33. Manley BFJ. Multivariate statistical methods. A primer. London: Chapman and Hall, 1986.Search in Google Scholar

34. Blagojevic D, Buzadzic B, Korac B, Saicic ZS, Radojicic R, Spasic MB, et al. Seasonal changes in the antioxidant defense in ground squirrel (Citellus citellus): possible role of GSH-Px. J Environ Pathol Toxicol Oncol 1998; 17:241–50.Search in Google Scholar

35. Jovanović-Galović A, Blagojević DP, Grubor-Lajšić G, Worland R, Spasić MB. Role of anti-oxidant defense during different stages of preadult life cycle in European corn borer (Ostrinia nubilalis, Hubn.): diapause and metamorphosis. Arch Insect Biochem Physiol 2004; 55:79–89.10.1002/arch.10126Search in Google Scholar

36. Perluigi M, Fai Poon H, Hensley K, Pierce WM, Klein JB, Calabrese V, et al. Proteomic analysis of 4-hydroxy-2-nonenal-modified proteins in G93A-SOD1 transgenic mice − a model of familial amyotrophic lateral sclerosis. Free Radic Biol Med 2005; 38:960–8.10.1016/j.freeradbiomed.2004.12.021Search in Google Scholar

37. Nikolić A, Blagojević D, Stević Z, Niketić V, Saičić ZS, Spasić MB. Activities of AD enzymes in the blood of ALS patients – base for use of anti-oxidants in the treatment of ALS. In: Proceedings of the 11th Biennial Meeting of the Society for Free Radical Research International, 2002. Bologna, Italy: Medimond srl-Monduzzi Editore, 2002:323–6.Search in Google Scholar

38. Mase G, Ros S, Gemma A, Bonfigli L, Carraro N, Cazzato G, et al. ALS with variable phenotypes in a six-generation family caused by leu144phe mutation in the SOD1 gene. J Neurol Sci 2001; 191:11–8.10.1016/S0022-510X(01)00625-6Search in Google Scholar

39. Oteiza PI, Uchitel OD, Carrasquedo F, Dubrovski AL, Roma JC, Fraga CG. Evaluation of anti-oxidants, protein, and lipid oxidation products in blood from sporadic amyotrophic lateral sclerosis patients. Neurochem Res 1997; 22:535–9.10.1023/A:1027384432715Search in Google Scholar

40. Dobashi K, Pahan K, Chahal A, Singh I. Modulation of endogenous antioxidant enzymes by nitric oxide in rat C6 glial cells. J Neurochem 1997; 68:1896–903.10.1046/j.1471-4159.1997.68051896.xSearch in Google Scholar PubMed

41. Kim YS, Han S. Nitric oxide protects Cu,Zn-superoxide dismutase from hydrogen peroxide-induced inactivation. FEBS Lett 2000; 479:25–8.10.1016/S0014-5793(00)01874-3Search in Google Scholar

42. Niketic V, Stojanovic S, Nikolic A, Spasic M, Michelson AM. Exposure of Mn and Fe SODs, but not Cu/Zn SOD, to NO leads to nitrosonium and nitroxyl ions generation which cause enzyme modification and inactivation: an in vitro study. Free Radic Biol Med 1999; 27:992–6.10.1016/S0891-5849(98)00256-1Search in Google Scholar

43. Poon HF, Hensley K, Thongboonkerd V, Merchant ML, Lynn BC, Pierce WM, et al. Redox proteomics analysis of oxidatively modified proteins in G93A-SOD1 transgenic mice − a model of familial amyotrophic lateral sclerosis. Free Radic Biol Med 2005; 39:453–62.10.1016/j.freeradbiomed.2005.03.030Search in Google Scholar PubMed

44. Bruijn LI, Miller TM, Cleveland DW. Unraveling the mechanisms involved in motor neuron degeneration in ALS. Annu Rev Neurosci 2004; 27:723–49.10.1146/annurev.neuro.27.070203.144244Search in Google Scholar PubMed

45. Pioro E. Antioxidant therapy in ALS. Amyotroph Lateral Scler Other Motor Neuron Disord 2000; 4:5–15.10.1080/14660820050515656Search in Google Scholar PubMed

Received: 2005-10-13
Accepted: 2006-2-20
Published Online: 2011-9-21
Published in Print: 2006-5-1

©2006 by Walter de Gruyter Berlin New York

Downloaded on 1.6.2024 from https://www.degruyter.com/document/doi/10.1515/CCLM.2006.111/html
Scroll to top button