A Cystine-Rich Whey Supplement (Immunocal®) Provides Neuroprotection from Diverse Oxidative Stress-Inducing Agents In Vitro by Preserving Cellular Glutathione

Aimee N Winter, Erika K Ross, Vamsi Daliparthi, Whitney A Sumner, Danielle M Kirchhof, Evan Manning, Heather M Wilkins, Daniel A Linseman, Aimee N Winter, Erika K Ross, Vamsi Daliparthi, Whitney A Sumner, Danielle M Kirchhof, Evan Manning, Heather M Wilkins, Daniel A Linseman

Abstract

Oxidative stress is a principal mechanism underlying the pathophysiology of neurodegeneration. Therefore, nutritional enhancement of endogenous antioxidant defenses may represent a viable treatment option. We investigated the neuroprotective properties of a unique whey protein supplement (Immunocal®) that provides an essential precursor (cystine) for synthesis of the endogenous antioxidant, glutathione (GSH). Primary cultures of rat cerebellar granule neurons (CGNs), NSC34 motor neuronal cells, or HT22 hippocampal cells were preincubated in medium containing Immunocal and then subsequently treated with agents known to induce oxidative stress. Immunocal protected CGNs against neurotoxicity induced by the Bcl-2 inhibitor, HA14-1, the nitric oxide donor, sodium nitroprusside, CuCl2, and AlCl3. Immunocal also significantly reduced NSC34 cell death due to either H2O2 or glutamate and mitigated toxicity in HT22 cells overexpressing β-amyloid1-42. The neuroprotective effects of Immunocal were blocked by inhibition of γ-glutamyl-cysteine ligase, demonstrating dependence on de novo GSH synthesis. These findings indicate that sustaining GSH with Immunocal significantly protects neurons against diverse inducers of oxidative stress. Thus, Immunocal is a nutritional supplement worthy of testing in preclinical animal models of neurodegeneration and in future clinical trials of patients afflicted by these diseases.

Figures

Figure 1
Figure 1
Cells treated with Immunocal display healthy neuronal morphology. Cells were left untreated (a) or treated with Immunocal alone (b) and assessed for overall health and appearance. Left-hand panels are representative images of cell nuclei stained with DAPI. Right-hand panels depict the same fields as viewed under brightfield to assess the state of neuronal processes and soma. Con: control; ICAL: Immunocal. Scale bar, 10 μm.
Figure 2
Figure 2
Immunocal preserves cellular GSH and prevents apoptosis in CGNs exposed to the Bcl-2 inhibitor, HA14-1. (a) Representative images of CGNs left untreated (control), treated with HA14-1 (15 μM), or preincubated for 24 h with Immunocal before HA14-1 treatment for further 24 h. Panels from left to right, DAPI (nuclei), β-tubulin, merged images showing β-tubulin (green), and DAPI (blue). Scale bar, 10 μm. (b) Quantification of apoptosis for 4 independent experiments performed as in (a) except some cultures were preincubated with 200 μM BSO as well. Apoptotic cells were those with condensed or fragmented nuclei. Results are shown as mean ± SEM, n = 4. ∗∗∗ indicates p < 0.001 compared to control, ††† indicates p < 0.001 compared to HA14-1, ‡‡‡ indicates p < 0.001 compared to ICAL + HA14-1. (c) CGNs were treated exactly as described in (b). Total cellular GSH was measured as described in Materials and Methods. Data shown represent the percent of control cellular GSH concentration, mean ± SEM, n = 4. ∗∗∗ indicates p < 0.001 compared to control, † indicates p < 0.05 compared to HA14-1, and ‡‡ indicates p < 0.01 compared to ICAL + HA14-1. Significant differences were determined by one-way ANOVA with a post hoc Tukey's test. Con: control; ICAL: Immunocal; BSO: buthionine sulfoximine.
Figure 3
Figure 3
Immunocal decreases CuCl2-induced apoptosis and lipid peroxidation in CGNs. (a) Representative images of CGNs left untreated (control), treated with CuCl2 (50 μM), or preincubated with Immunocal for 24 h before CuCl2 treatment for further 24 h. Immunofluorescence shows β-tubulin (green) and DAPI (blue). Scale bar, 10 μm. (b) Quantification of apoptosis for 4 independent experiments performed as in (a). Results are shown as mean ± SEM, n = 4. ∗∗ indicates p < 0.01 compared to control and †† indicates p < 0.01 compared to CuCl2. (c) Cellular lipid peroxidation (malondialdehyde (MDA)) was measured as described in Materials and Methods. Results are shown as mean ± SEM, n = 5. ∗∗ indicates p < 0.01 compared to control, ††† indicates p < 0.001 compared to CuCl2. Con: control; ICAL: Immunocal.
Figure 4
Figure 4
Immunocal preserves CGN viability and protects from apoptosis after exposure to SNP. (a) Representative images of CGNs left untreated (control), treated with SNP (100 μM), or preincubated with Immunocal for 24 h before SNP treatment for further 24 h. Immunofluorescence shows β-tubulin (green) and DAPI (blue). Scale bar, 10 μm. (b) Quantification of apoptosis for 5 independent experiments performed as in (a). Results are shown as mean ± SEM, n = 5. (c) MTT cell viability was measured as described in Materials and Methods. Results are shown as mean ± SEM, n = 3. For (b) and (c), ∗∗∗ indicates p < 0.001 compared to control, and ††† indicates p < 0.001 compared to SNP. Con: control; ICAL: Immunocal.
Figure 5
Figure 5
Immunocal protects CGNs from AlCl3-induced toxicity. (a) Representative images of CGNs left untreated (control), treated with AlCl3 (10 μM), or preincubated with Immunocal for 24 h before AlCl3 treatment for further 48 h. Panels from left to right, DAPI (nuclei), β-tubulin, and merged image showing β-tubulin (green), and DAPI (blue). Scale bar, 10 μm. (b) CGN apoptosis was quantified for 4 independent experiments as described in (a). Results are shown as mean ± SEM, n = 4. ∗∗∗ indicates p < 0.001 compared to control, and †† indicates p < 0.01 compared to AlCl3. Con: control; ICAL: Immunocal.
Figure 6
Figure 6
Immunocal protects NSC34 cells from H2O2 and glutamate/glycine-induced excitotoxicity. (a) Cell survival was quantified with MTT cell viability assay for 5 independent experiments in undifferentiated NSC34 left untreated (control), treated with H2O2 (250 μM), or preincubated with Immunocal for 24 h before H2O2 treatment for further 24 h. Results are shown as mean ± SEM, n = 5. ∗∗ indicates p < 0.01 compared to control, † indicates p < 0.05 compared to H2O2, and †† indicates p < 0.01 compared to H2O2. (b) Representative images showing morphological differences between undifferentiated (wildtype (WT)) and differentiated (DIFF) NSC34 cells, β-tubulin (green), and DAPI (blue). Scale bar, 10 μm. (c) Cell survival was quantified for 5 independent experiments with an MTT cell viability assay in differentiated NSC34 cells left untreated (control), treated with glutamate/glycine (1 mM/100 μM), or preincubated with Immunocal for 24 h before glutamate/glycine treatment for further 24 h. ∗ indicates p < 0.05 compared to control, and † indicates p < 0.05 compared to glutamate/glycine. Con: control; ICAL: Immunocal; GG: glutamate/glycine.
Figure 7
Figure 7
Immunocal protects HT22 cells from toxicity induced by overexpression of Aβ1-42. (a) Representative images of HT22 cells transfected with either empty vector (IRES) or Aβ1-42. Top panels display colored images showing successful transfection of the cells, and bottom panels display decolorized images of cell nuclei to visualize nuclear condensation. Arrows indicate transfected cells. (b) Quantification of apoptosis for 4 independent experiments performed as in (a). Results are shown as mean ± SEM, n = 4. ∗∗∗ indicates p < 0.001 compared to control, and ††† indicates p < 0.001 compared to cells transfected with Aβ1-42 without Immunocal preincubation. Aβ: amyloid-beta; ICAL: Immunocal.
Figure 8
Figure 8
Proposed neuroprotective mechanism of Immunocal. Immunocal provides the essential GSH precursor, cystine, which is transported into cerebellar granule neurons via the system xc− antiporter (Sxc−). Upon entry into the cell, cystine is rapidly hydrolyzed to form two cysteine molecules, which are then utilized in the de novo synthesis of GSH by γ-glutamylcysteine ligase (γ-GCL) and glutathione synthase (GSS). Newly synthesized glutathione inhibits oxidation caused by a variety of insults, thereby preventing mitochondrial oxidative stress (MOS) and subsequent induction of apoptosis.

References

    1. Lin M. T., Beal M. F. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. 2006;443:787–795. doi: 10.1038/nature05292.
    1. Smith E. F., Shaw P. J., De Vos K. J. The role of mitochondria in amyotrophic lateral sclerosis. Neuroscience Letters. 2017 doi: 10.1016/j.neulet.2017.06.052.
    1. Cassano T., Pace L., Bedse G., et al. Glutamate and mitochondria: two prominent players in the oxidative stress-induced neurodegeneration. Current Alzheimer Research. 2016;13:185–197. doi: 10.2174/1567205013666151218132725.
    1. Jiang T., Sun Q., Chen S. Oxidative stress: a major pathogenesis and potential therapeutic target of antioxidative agents in Parkinson’s disease and Alzheimer’s disease. Progress in Neurobiology. 2016;147:1–19. doi: 10.1016/j.pneurobio.2016.07.005.
    1. Dawson T. M., Dawson V. L. Molecular pathways of neurodegeneration in Parkinson’s disease. Science. 2003;302:819–822. doi: 10.1126/science.1087753.
    1. Zuo L., Motherwell M. S. The impact of reactive oxygen species and genetic mitochondrial mutations in Parkinson’s disease. Gene. 2013;532:18–23. doi: 10.1016/j.gene.2013.07.085.
    1. Nunomura A., Perry G., Aliev G., et al. Oxidative damage is the earliest event in Alzheimer disease. Journal of Neuropathology & Experimental Neurology. 2001;60:759–767. doi: 10.1093/jnen/60.8.759.
    1. Leuner K., Müller W. E., Reichert A. S. From mitochondrial dysfunction to amyloid beta formation: novel insights into the pathogenesis of Alzheimer’s disease. Molecular Neurobiology. 2012;46:186–193. doi: 10.1007/s12035-012-8307-4.
    1. Ferri A., Cozzolino M., Crosio C., et al. Familial ALS-superoxide dismutases associate with mitochondria and shift their redox potentials. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:13860–13865. doi: 10.1073/pnas.0605814103.
    1. Franco R., Cidlowski J. A. Apoptosis and glutathione: beyond an antioxidant. Cell Death and Differentiation. 2009;16:1303–1314. doi: 10.1038/cdd.2009.107.
    1. Babu G. N., Kumar A., Chandra R., et al. Oxidant-antioxidant imbalance in the erythrocytes of sporadic amyotrophic lateral sclerosis patients correlates with the progression of disease. Neurochemistry International. 2008;52:1284–1289. doi: 10.1016/j.neuint.2008.01.009.
    1. Calabrese V., Sultana R., Scapagnini G., et al. Nitrosative stress, cellular stress response and thiol homeostasis in patients with Alzheimer’s disease. Antioxidants & Redox Signaling. 2006;8:1975–1986. doi: 10.1089/ars.2006.8.1975.
    1. Pearce R. K. B., Owen A., Daniel S., Jenner P., Marsden C. D. Alterations in the distribution of glutathione in the substantia nigra in Parkinson’s disease. Journal of Neural Transmission. 1997;104:661–677. doi: 10.1007/BF01291884.
    1. Garrido M., Tereshchenko Y., Zhevtsova Z. Glutathione depletion and overproduction both initiate degeneration of nigral dopaminergic neurons. Acta Neuropathologica. 2011;121:475–485. doi: 10.1007/s00401-010-0791-x.
    1. Hsu M., Srinivas B., Kumar J., Subramanian R., Andersen J. Glutathione depletion resulting in selective mitochondrial complex I inhibition in dopaminergic cells is via an NO-mediated pathway not involving peroxynitrite: implications for Parkinson’s disease. Journal of Neurochemistry. 2005;92:1091–1103. doi: 10.1111/j.1471-4159.2004.02929.x.
    1. Muyderman H., Hutson P. G., Matusica D., Rogers M. L., Rush R. A. The human G93A-superoxide dismutase-1 mutation, mitochondrial glutathione and apoptotic cell death. Neurochemical Research. 2009;34:1847–1856. doi: 10.1007/s11064-009-9974-z.
    1. Du T., Ciccotosto G. D., Cranston G. A., et al. Neurotoxicity from glutathione depletion is mediated by Cu-dependent p53 activation. Free Radical Biology and Medicine. 2008;44:44–55. doi: 10.1016/j.freeradbiomed.2007.09.001.
    1. Iguchi Y., Katsuno M., Takagi S., et al. Oxidative stress induced by glutathione depletion reproduces pathological modifications of TDP-43 linked to TDP-43 proteinopathies. Neurobiology of Disease. 2012;45:862–870. doi: 10.1016/j.nbd.2011.12.002.
    1. Jaiswal A. K. Nrf2 signaling in coordinated activation of antioxidant gene expression. Free Radical Biology and Medicine. 2004;36:1199–1207. doi: 10.1016/j.freeradbiomed.2004.02.074.
    1. Sun L., Gu L., Wang S., Yuan J., Yang H., Zhu J. N-acetylcysteine protects against apoptosis through modulation of group I metabotropic glutamate receptor activity. PLoS One. 2012;7, article e32503 doi: 10.1371/journal.pone.0032503.
    1. Andreassen O. A., Dedeoglu A., Klivenyi P., Beal M. F., Bush A. I. N-acetyl-L-cysteine improves survival and preserves motor performance in an animal model of familial amyotrophic lateral sclerosis. Neuroreport. 2000;11:2491–2493. doi: 10.1097/00001756-200008030-00029.
    1. Farr S. A., Fai Poon H., Dogrukol-Ak D., et al. The antioxidants α-lipoic acid and N-acetylcysteine reverse memory impairment and brain oxidative stress in aged SAMP8 mice. Journal of Neurochemistry. 2003;84:1173–1183. doi: 10.1046/j.1471-4159.2003.01580.x.
    1. Zeevalk G. D., Manzino L., Sonsalla P. K., Bernard L. P. Characterization of intracellular elevation of glutathione (GSH) with glutathione monoethyl ester and GSH in brain and neuronal cultures: relevance to Parkinson’s disease. Experimental Neurology. 2007;203:512–520. doi: 10.1016/j.expneurol.2006.09.004.
    1. Mischley L. K., Leverenz J. B., Lau R. C., et al. A randomized, double-blind phase I/IIa study of intranasal glutathione in Parkinson’s disease. Movement Disorders. 2015;30:1696–1701. doi: 10.1002/mds.26351.
    1. Chen P. C., Vargas M. R., Pani A. K., et al. Nrf2-mediated neuroprotection in the MPTP mouse model of Parkinson’s disease: critical role for the astrocyte. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:2933–2938. doi: 10.1073/pnas.0813361106.
    1. Vargas M. R., Johnson D. A., Sirkis D. W., Messing A., Johnson J. A. Nrf2 activation in astrocytes protects against neurodegeneration in mouse models of familial amyotrophic lateral sclerosis. The Journal of Neuroscience. 2008;28:13574–13581. doi: 10.1523/JNEUROSCI.4099-08.2008.
    1. Neymotin A., Calingasan N. Y., Wille E., et al. Neuroprotective effect of Nrf2/ARE activators, CDDO ethylamide and CDDO trifluoroethylamide, in a mouse model of amyotrophic lateral sclerosis. Free Radical Biology and Medicine. 2011;51:88–96. doi: 10.1016/j.freeradbiomed.2011.03.027.
    1. Kanninen K., Heikkinen R., Malm T., et al. Intrahippocampal injection of a lentiviral vector expressing Nrf2 improves spatial learning in a mouse model of Alzheimer’s disease. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:16505–16510. doi: 10.1073/pnas.0908397106.
    1. Olsson B., Johansson M., Gabrielsson J., Bolme P. Pharmacokinetics and bioavailability of reduced and oxidized N-acetylcysteine. European Journal of Clinical Pharmacology. 1988;34:77–82. doi: 10.1007/BF01061422.
    1. Tsai W. Y., Chang W., Chen C., Lu F. Enhancing effect of patented whey protein isolate (Immunocal) on cytotoxicity of an anti-cancer drug. Nutrition and Cancer. 2000;38:200–208. doi: 10.1207/S15327914NC382_9.
    1. Micke P., Beeh K. M., Buhl R. Effects of long-term supplementation with whey proteins on plasma glutathione levels of HIV-infected patients. European Journal of Nutrition. 2002;41:12–18. doi: 10.1007/s003940200001.
    1. Grey V., Mohammed S. R., Smountasm A. A., Bahlool R., Lands L. C. Improved glutathione status in young adult patients with cystic fibrosis supplemented with whey protein. Journal of Cystic Fibrosis. 2003;2:195–198. doi: 10.1016/S1569-1993(03)00097-3.
    1. Bounous G., Gold P. The biological activity of undenatured dietary whey proteins: role of glutathione. Clinical and Investigative Medicine. 1991;14:296–309.
    1. Linseman D. A., Laessig T., Meintzer M. K., et al. An essential role for Rac/Cdc42 GTPases in cerebellar granule neuron survival. The Journal of Biological Chemistry. 2001;276:39123–39131. doi: 10.1074/jbc.M103959200.
    1. Baruchel S., Viau G. In vitro selective modulation of cellular glutathione by a humanized native milk protein isolate in normal cells and rat mammary carcinoma model. Anticancer Research. 1996;16:1095–1100.
    1. Baruchel S., Viau G., Olivier R., Bounous G., Wainberg M. A. Nutraceutical modulation of glutathione with a humanized native milk serum protein isolate, Immunocal®: application in AIDS and cancer. In: Montagnier L., Olivier R., Pasquier C., editors. Oxidative Stress in Cancer, AIDS, and Neurodegenerative Diseases. 1, Chapter 42. New York, NY, USA: Marcel Dekker, Inc.; 1998. pp. 447–462.
    1. Zimmermann A. K., Loucks F. A., Schroeder E. K., Bouchard R. J., Tyler K. L., Linseman D. A. Glutathione binding to the Bcl-2 homology-3 domain groove: a molecular basis for Bcl-2 antioxidant function at mitochondria. The Journal of Biological Chemistry. 2007;282:29296–29304. doi: 10.1074/jbc.M702853200.
    1. Wilkins H. M., Marquardt K., Lash L. H., Linseman D. A. Bcl-2 is a novel interacting partner for the 2-oxyglutarate carrier and a key regulator of mitochondrial glutathione. Free Radical Biology and Medicine. 2012;52:410–419. doi: 10.1016/j.freeradbiomed.2011.10.495.
    1. Drew R., Miners J. O. The effects of buthionine sulphoximine (BSO) on glutathione depletion and xenobiotic biotransformation. Biochemical Pharmacology. 1984;33:2989–2994. doi: 10.1016/0006-2952(84)90598-7.
    1. Arciello M., Rotilio G., Rossi L. Copper-dependent toxicity in SH-SY5Y neuroblastoma cells involves mitochondrial damage. Biochemical and Biophysical Research Communications. 2005;327:454–459. doi: 10.1016/j.bbrc.2004.12.022.
    1. Yurkova I. L., Arnhold J., Fitzl G., Huster D. Fragmentation of mitochondrial cardiolipin by copper ions in the Atp7b−/− mouse model of Wilson’s disease. Chemistry and Physics of Lipids. 2011;164:393–400. doi: 10.1016/j.chemphyslip.2011.05.006.
    1. Fukushima T., Koide M., Ago Y., Baba A., Matsuda T. T-817MA, a novel neurotrophic agent, improves sodium nitroprusside-induced mitochondrial dysfunction in cortical neurons. Neurochemistry International. 2006;48:124–130. doi: 10.1016/j.neuint.2005.08.012.
    1. Wei T., Chen C., Hou J., Zhao B., Xin W., Mori A. The antioxidant EPC-K1 attenuates NO-induced mitochondrial dysfunction, lipid peroxidation and apoptosis in cerebellar granule cells. Toxicology. 1999;134:117–126. doi: 10.1016/S0300-483X(99)00030-X.
    1. Kumar V., Gill K. D. Aluminum neurotoxicity: neurobehavioural and oxidative aspects. Archives of Toxicology. 2009;83:965–978. doi: 10.1007/s00204-009-0455-6.
    1. Niu P. Y., Niu Q., Zhang Q. L., et al. Aluminum impairs rat neural cell mitochondria in vitro. International Journal of Immunopathology and Pharmacology. 2005;18:683–689. doi: 10.1177/039463200501800410.
    1. Whittemore E. R., Loo D. T., Watt J. A., Cotman C. W. A detailed analysis of hydrogen peroxide-induced cell death in primary neuronal culture. Neuroscience. 1995;67:921–932. doi: 10.1016/0306-4522(95)00108-U.
    1. Eggett C. J., Crosier S., Manning P., et al. Development and characterization of a glutamate-sensitive motor neuron cell line. Journal of Neurochemistry. 2000;74:1895–1902. doi: 10.1046/j.1471-4159.2000.0741895.x.
    1. Mao P., Reddy P. H. Aging and amyloid beta-induced oxidative DNA damage and mitochondrial dysfunction in Alzheimer’s disease: implications for early intervention and therapeutics. Biochimica et Biophysica Acta. 2011;1812:1359–1370. doi: 10.1016/j.bbadis.2011.08.005.
    1. Bounous G., Baruchel S. Whey proteins as a food supplement in HIV-seropositive individuals. Clinical and Investigative Medicine. 1993;16:204–209.
    1. Jang J. H., Surh Y. J. Bcl-2 attenuation of oxidative cell death is associated with upregulation of gamma-glutamylcysteine ligase via constitutive NF-kappaB activation. The Journal of Biological Chemistry. 2004;279:38779–38786. doi: 10.1074/jbc.M406371200.
    1. Meredith M. J., Cusick C. L., Soltninassab S., Sekhar K. S., Lu S., Freeman M. L. Expression of Bcl-2 increases intracellular glutathione by inhibiting methionine-dependent GSH efflux. Biochemical and Biophysical Research Communications. 1998;248:458–463. doi: 10.1006/bbrc.1998.8998.
    1. Hochman A., Stemin H., Gorodin S., et al. Enhanced oxidative stress and altered antioxidants in brains of Bcl-2 deficient mice. Journal of Neurochemistry. 1998;71:741–748. doi: 10.1046/j.1471-4159.1998.71020741.x.
    1. Freedman J. H., Ciriolo M., Peisach J. The role of glutathione in copper metabolism and toxicity. The Journal of Biological Chemistry. 1989;264:5596–5605.
    1. Sayre L. M., Perry G., Harris P. L., Liu Y., Schubert K. A., Smith M. A. In situ oxidative catalysis by neurofibrillary tangles and senile plaques in Alzheimer’s disease: a central role for bound transition metals. Journal of Neurochemistry. 2000;74:270–279. doi: 10.1046/j.1471-4159.2000.0740270.x.
    1. White A. R., Cappai R. Neurotoxicity from glutathione depletion is dependent on extracellular trace copper. Journal of Neuroscience Research. 2003;71:889–897. doi: 10.1002/jnr.10537.
    1. Trumbull K. A., Beckman J. S. A role for copper in the toxicity of zinc-deficient superoxide dismutase to motor neurons in amyotrophic lateral sclerosis. Antioxidants & Redox Signaling. 2009;11:1627–1639. doi: 10.1089/ars.2009.2574.
    1. Di Filippo M., Chisserini D., Tozzi A., Picconi B., Calabresi P. Mitochondria and the link between neuroinflammation and neurodegeneration. Journal of Alzheimer's Disease. 2010;20:S369–S379. doi: 10.3233/JAD-2010-100543.
    1. Yuste J. E., Tarragon E., Campuzano C. M., Ros-Bernal F. Implications of glial nitric oxide in neurodegenerative diseases. Frontiers in Cellular Neuroscience. 2015;9:p. 322. doi: 10.3389/fncel.2015.00322.
    1. Dias C., Lourenco C. F., Ferreiro E., Barbosa R. M., Laranjinha J., Ledo A. Age-dependent changes in the glutamate-nitric oxide pathway in the hippocampus of the triple transgenic model of Alzheimer’s disease: implications for neurometabolic regulation. Neurobiology of Aging. 2016;46:84–95. doi: 10.1016/j.neurobiolaging.2016.06.012.
    1. Fernández E., Garcia-Moreno J. M., Martin de Pablos A., Chacón J. May the evaluation of nitrosative stress through selective increase of 3-nitrotyrosie proteins other than nitroalbumin and dominant tyrosine-125/136 nitrosylation of serum α-synuclein serve for diagnosis of sporadic Parkinson’s disease? Antioxidants & Redox Signaling. 2013;19:912–918. doi: 10.1089/ars.2013.5250.
    1. Murakami K., Yoshino M. Aluminum decreases the glutathione regeneration by the inhibition of NADP-iscocitrate dehydrogenase in mitochondria. Journal of Cellular Biochemistry. 2004;93:1267–1271. doi: 10.1002/jcb.20261.
    1. Shaw C., Petrik M. S. Aluminum hydroxide injections lead to motor deficits and motor neuron degeneration. Journal of Inorganic Biochemistry. 2009;103:1555–1562. doi: 10.1016/j.jinorgbio.2009.05.019.
    1. Haley R. W. Excess incidence of ALS in young Gulf War veterans. Neurology. 2003;61:750–756. doi: 10.1212/WNL.61.6.750.
    1. Cashman N. R., Durham H. D., Blusztain J. K., et al. Neuroblastoma x spinal cord (NSC) hybrid cell lines resemble developing motor neurons. Developmental Dynamics. 1992;194:209–221. doi: 10.1002/aja.1001940306.
    1. Lewerenz J., Maher P. Chronic glutamate toxicity in neurodegenerative diseases – what is the evidence? Frontiers in Neuroscience. 2015;9:p. 469. doi: 10.3389/fnins.2015.00469.
    1. Caspersen C., Wang N., Yao J., et al. Mitochondrial Abeta: a potential focal point for neuronal metabolic dysfunction in Alzheimer’s disease. The FASEB Journal. 2005;19:2040–2041. doi: 10.1096/fj.05-3735fje.
    1. Manczak M., Calkins M. J., Reddy P. H. Impaired mitochondrial dynamics and abnormal interaction of amyloid beta with mitochondrial protein Drp1 in neurons from patients with Alzheimer’s disease: implications for neuronal damage. Human Molecular Genetics. 2011;20:2495–2509. doi: 10.1093/hmg/ddr139.
    1. Walls K. C., Coskun P., Gallegos-Perez J. L., et al. Swedish Alzheimer mutation induces mitochondrial dysfunction mediated by HSP60 mislocalization of amyloid precursor protein (APP) and beta-amyloid. The Journal of Biological Chemistry. 2012;287:30317–30327. doi: 10.1074/jbc.M112.365890.
    1. Song W., Tavitian A., Cressatti M., Galindez C., Liberman A., Schipper H. M. Cysteine-rich whey protein isolate (Immunocal®) ameliorates deficits in the GFAP.HMOX1 mouse model of schizophrenia. Free Radical Biology and Medicine. 2017;110:162–175. doi: 10.1016/j.freeradbiomed.2017.05.025.
    1. Ross E. K., Winter A. N., Wilkins H. M., et al. A cystine-rich whey supplement (Immunocal®) delays disease onset and prevents spinal cord glutathione depletion in the hSOD1G93A mouse model of amyotrophic lateral sclerosis. Antioxidants. 2014;3:843–865. doi: 10.3390/antiox3040843.

Source: PubMed

3
Abonnere