Generic placeholder image

Current Aging Science

Editor-in-Chief

ISSN (Print): 1874-6098
ISSN (Online): 1874-6128

Review Article

Oxidative and Inflammatory Events in Prion Diseases: Can They Be Therapeutic Targets?

Author(s): Kedar N. Prasad* and Stephen C. Bondy

Volume 11, Issue 4, 2018

Page: [216 - 225] Pages: 10

DOI: 10.2174/1874609812666190111100205

Abstract

Prion diseases are a group of incurable infectious terminal neurodegenerative diseases caused by the aggregated misfolded PrPsc in selected mammals including humans. The complex physical interaction between normal prion protein PrPc and infectious PrPsc causes conformational change from the α- helix structure of PrPc to the β-sheet structure of PrPsc, and this process is repeated. Increased oxidative stress is one of the factors that facilitate the conversion of PrPc to PrPsc. This overview presents evidence to show that increased oxidative stress and inflammation are involved in the progression of this disease. Evidence is given for the participation of redoxsensitive metals Cu and Fe with PrPsc inducing oxidative stress by disturbing the homeostasis of these metals. The fact that some antioxidants block the toxicity of misfolded PrPc peptide supports the role of oxidative stress in prion disease. After exogenous infection in mice, PrPsc enters the follicular dendritic cells where PrPsc replicates before neuroinvasion where they continue to replicate and cause inflammation leading to neurodegeneration. Therefore, reducing levels of oxidative stress and inflammation may decrease the rate of the progression of this disease. It may be an important order to reduce oxidative stress and inflammation at the same time. This may be achieved by increasing the levels of antioxidant enzymes by activating the Nrf2 pathway together with simultaneous administration of dietary and endogenous antioxidants. It is proposed that a mixture of micronutrients could enable these concurrent events thereby reducing the progression of human prion disease.

Keywords: Oxidative stress, inflammation, apoptosis, antioxidants, misfolded proteins, spongiform encephalopathy, prion diseases.

Graphical Abstract
[1]
Gajdusek DC, Zigas V. Degenerative disease of the central nervous system in New Guinea; the endemic occurrence of kuru in the native population. N Engl J Med 1957; 257(20): 974-8.
[2]
Gibbs CJ, Gajdusek DC, Asher DM, et al. Creutzfeldt-Jakob disease (spongiform encephalopathy): Transmission to the chimpanzee. Science 1968; 161(3839): 388-9.
[3]
Prusiner SB. Novel proteinaceous infectious particles cause scrapie. Science 1982; 216(4542): 136-44.
[4]
Prusiner SB. Cell biology. A unifying role for prions in neurodegenerative diseases. Science 2012; 336(6088): 1511-3.
[5]
Chen YR, Yi FF, Li XY, et al. Resveratrol attenuates ventricular arrhythmias and improves the long-term survival in rats with myocardial infarction. Cardiovasc Drugs Ther 2008; 22(6): 479-85.
[6]
Brazier MW, Lewis V, Ciccotosto GD, et al. Correlative studies support lipid peroxidation is linked to PrP(res) propagation as an early primary pathogenic event in prion disease. Brain Res Bull 2006; 68(5): 346-54.
[7]
Crespo I, Roomp K, Jurkowski W, et al. Gene regulatory network analysis supports inflammation as a key neurodegeneration process in prion disease. BMC Syst Biol 2012; 6: 132.
[8]
Algarzae N, Hebron M, Miessau M, et al. Parkin prevents cortical atrophy and Abeta-induced alterations of brain metabolism: (1)(3)C NMR and magnetic resonance imaging studies in AD models. Neuroscience 2012; 225: 22-34.
[9]
Carroll JA, Striebel JF, Race B, et al. Prion infection of mouse brain reveals multiple new upregulated genes involved in neuroinflammation or signal transduction. J Virol 2015; 89(4): 2388-404.
[10]
Mustafa AG, Singh IN, Wang J, et al. Mitochondrial protection after traumatic brain injury by scavenging lipid peroxyl radicals. J Neurochem 2010; 114(1): 271-80.
[11]
Yen CF, Harischandra DS, Kanthasamy A, et al. Copper-induced structural conversion templates prion protein oligomerization and neurotoxicity. Sci Adv 2016; 2(7): e1600014.
[12]
Asuni AA, Guridi M, Sanchez S, et al. Antioxidant peroxiredoxin 6 protein rescues toxicity due to oxidative stress and cellular hypoxia in vitro, and attenuates prion-related pathology in vivo. Neurochem Int 2015; 90: 152-65.
[13]
Lee ST, Chu K, Jung KH, et al. Altered expression of miR-202 in cerebellum of multiple-system atrophy. Mol Neurobiol 2015; 51(1): 180-6.
[14]
Mizrahi M, Friedman-Levi Y, Larush L, et al. Pomegranate seed oil nanoemulsions for the prevention and treatment of neurodegenerative diseases: The case of genetic CJD. Nanomedicine 2014; 10(6): 1353-63.
[15]
Brazier MW, Doctrow SR, Masters CL, et al. A manganese-superoxide dismutase/catalase mimetic extends survival in a mouse model of human prion disease. Free Radic Biol Med 2008; 45(2): 184-92.
[16]
Holman RC, Belay ED, Christensen KY, et al. Human prion diseases in the United States. PLoS One 2010; 5(1): e8521.
[17]
Parchi P, Giese A, Capellari S, et al. Classification of sporadic Creutzfeldt-Jakob disease based on molecular and phenotypic analysis of 300 subjects. Ann Neurol 1999; 46(2): 224-33.
[18]
Hsiao K, Meiner Z, Kahana E, et al. Mutation of the prion protein in Libyan Jews with Creutzfeldt-Jakob disease. N Engl J Med 1991; 324(16): 1091-7.
[19]
Meiner Z, Gabizon R, Prusiner SB. Familial Creutzfeldt-Jakob disease. Codon 200 prion disease in Libyan Jews. Medicine (Baltimore) 1997; 76(4): 227-37.
[20]
Belay ED, Sejvar JJ, Shieh WJ, et al. Variant Creutzfeldt-Jakob disease death, United States. Emerg Infect Dis 2005; 11(9): 1351-4.
[21]
Ghosh S. Mechanism of intestinal entry of infectious prion protein in the pathogenesis of variant Creutzfeldt-Jakob disease. Adv Drug Deliv Rev 2004; 56(6): 915-20.
[22]
Belay ED. Transmissible spongiform encephalopathies in humans. Annu Rev Microbiol 1999; 53: 283-314.
[23]
Valleron AJ, Boelle PY, Will R, et al. Estimation of epidemic size and incubation time based on age characteristics of vCJD in the United Kingdom. Science 2001; 294(5547): 1726-8.
[24]
Armstrong RA, Cairrns NJ, Lantos PL. Quantification of the vacuolation (spongiform change) and prion protein deposition in 11 patients with s[oradic Creutzfeldt-Jakob disease. Acta Neuropathol 2001; 102: 591-6.
[25]
Iwasaki Y. Creutzfeldt-Jakob disease. Neuropathology 2017; 37(2): 174-88.
[26]
Bradford BM, Crocker PR, Mabbott NA. Peripheral prion disease pathogenesis is unaltered in the absence of sialoadhesin (Siglec-1/CD169). Immunology 2014; 143(1): 120-9.
[27]
Prinz M, Heikenwalder M, Junt T, et al. Positioning of follicular dendritic cells within the spleen controls prion neuroinvasion. Nature 2003; 425(6961): 957-62.
[28]
Prinz M, Montrasio F, Klein MA, et al. Lymph nodal prion replication and neuroinvasion in mice devoid of follicular dendritic cells. Proc Natl Acad Sci USA 2002; 99(2): 919-24.
[29]
Grassmann A, Wolf H, Hofmann J, et al. Cellular aspects of prion replication in vitro. Viruses 2013; 5(1): 374-405.
[30]
Basler K, Oesch B, Scott M, et al. Scrapie and cellular PrP isoforms are encoded by the same chromosomal gene. Cell 1986; 46(3): 417-28.
[31]
Chesebro B, Race R, Wehrly K, et al. Identification of scrapie prion protein-specific mRNA in scrapie-infected and uninfected brain. Nature 1985; 315(6017): 331-3.
[32]
Rigter A, Priem J, Timmers-Parohi D, et al. Prion protein self-peptides modulate prion interactions and conversion. BMC Biochem 2009; 10: 29.
[33]
Ashok A, Hegde RS. Selective processing and metabolism of disease-causing mutant prion proteins. PLoS Pathog 2009; 5(6): e1000479.
[34]
Bossers A, Belt P, Raymond GJ, et al. Scrapie susceptibility-linked polymorphisms modulate the in vitro conversion of sheep prion protein to protease-resistant forms. Proc Natl Acad Sci USA 1997; 94(10): 4931-6.
[35]
Sabuncu E, Petit S, Le Dur A, et al. PrP polymorphisms tightly control sheep prion replication in cultured cells. J Virol 2003; 77(4): 2696-700.
[36]
Kobayashi A, Teruya K, Matsuura Y, et al. The influence of PRNP polymorphisms on human prion disease susceptibility: An update. Acta Neuropathol 2015; 130(2): 159-70.
[37]
Apetri AC, Vanik DL, Surewicz WK. Polymorphism at residue 129 modulates the conformational conversion of the D178N variant of human prion protein 90-231. Biochemistry 2005; 44(48): 15880-8.
[38]
Nadal RC, Abdelraheim SR, Brazier MW, et al. Prion protein does not redox-silence Cu2+, but is a sacrificial quencher of hydroxyl radicals. Free Radic Biol Med 2007; 42(1): 79-89.
[39]
Basu S, Mohan ML, Luo X, et al. Modulation of proteinase K-resistant prion protein in cells and infectious brain homogenate by redox iron: Implications for prion replication and disease pathogenesis. Mol Biol Cell 2007; 18(9): 3302-12.
[40]
Kretzschmar HA, Tings T, Madlung A, et al. Function of PrP(C) as a copper-binding protein at the synapse. Arch Virol Suppl 2000; 16: 239-49.
[41]
Millhauser GL. Copper and the prion protein: Methods, structures, function, and disease. Annu Rev Phys Chem 2007; 58: 299-320.
[42]
Kamboj SS, Chopra K, Sandhir R. Hyperglycemia-induced alterations in synaptosomal membrane fluidity and activity of membrane bound enzymes: Beneficial effect of N-acetylcysteine supplementation. Neuroscience 2009; 162(2): 349-58.
[43]
Kawano T. Prion-derived copper-binding peptide fragments catalyze the generation of superoxide anion in the presence of aromatic monoamines. Int J Biol Sci 2007; 3: 57-63.
[44]
Aarsland D, Rongve A, Nore SP, et al. Frequency and case identification of dementia with Lewy bodies using the revised consensus criteria. Dement Geriatr Cogn Disord 2008; 26(5): 445-52.
[45]
Vilette D, Laulagnier K, Huor A, et al. Efficient inhibition of infectious prions multiplication and release by targeting the exosomal pathway. Cell Mol Life Sci 2015; 72(22): 4409-27.
[46]
Guo BB, Bellingham SA, Hill AF. Stimulating the release of exosomes increases the intercellular transfer of prions. J Biol Chem 2016; 291(10): 5128-37.
[47]
Lastres-Becker I. Role of the transcription factor Nrf2 in Parkinson’s disease: New Insights. J Alzheimers Dis Parkinsonism 2017; 7(4): 1-9.
[48]
Vella LJ, Sharples RA, Lawson VA, et al. Packaging of prions into exosomes is associated with a novel pathway of PrP processing. J Pathol 2007; 211(5): 582-90.
[49]
Fevrier B, Vilette D, Archer F, et al. Cells release prions in association with exosomes. Proc Natl Acad Sci USA 2004; 101(26): 9683-8.
[50]
Mays CE, Ryou C. Plasminogen: A cellular protein cofactor for PrPSc propagation. Prion 2011; 5(1): 22-7.
[51]
Kovacs GG, Seguin J, Quadrio I, et al. Genetic Creutzfeldt-Jakob disease associated with the E200K mutation: Characterization of a complex proteinopathy. Acta Neuropathol 2011; 121(1): 39-57.
[52]
Chen PY, Hou CW, Shibu MA, et al. Protective effect of Co-enzyme Q10 On doxorubicin-induced cardiomyopathy of rat hearts. Environ Toxicol 2017; 32(2): 679-89.
[53]
Arlt S, Kontush A, Zerr I, et al. Increased lipid peroxidation in cerebrospinal fluid and plasma from patients with Creutzfeldt-Jakob disease. Neurobiol Dis 2002; 10(2): 150-6.
[54]
Younan ND, Nadal RC, Davies P, et al. Methionine oxidation perturbs the structural core of the prion protein and suggests a generic misfolding pathway. J Biol Chem 2012; 287(34): 28263-75.
[55]
Brown DR. Neurodegeneration and oxidative stress: Prion disease results from loss of antioxidant defence. Folia Neuropathol 2005; 43(4): 229-43.
[56]
Che H, Sun LH, Guo F, et al. Expression of amyloid-associated miRNAs in both the forebrain cortex and hippocampus of middle-aged rat. Cell Physiol Biochem 2014; 33(1): 11-22.
[57]
Canello T, Frid K, Gabizon R, et al. Oxidation of Helix-3 methionines precedes the formation of PK resistant PrP. PLoS Pathog 2010; 6(7): e1000977.
[58]
Colombo G, Meli M, Morra G, et al. Methionine sulfoxides on prion protein Helix-3 switch on the alpha-fold destabilization required for conversion. PLoS One 2009; 4(1): e4296.
[59]
Coleman BM, Harrison CF, Guo B, et al. Pathogenic mutations within the hydrophobic domain of the prion protein lead to the formation of protease-sensitive prion species with increased lethality. J Virol 2014; 88(5): 2690-703.
[60]
Obst J, Simon E, Mancuso R, et al. The role of microglia in prion diseases: A paradigm of functional diversity. Front Aging Neurosci 2017; 9: 207.
[61]
Carroll JA, Striebel JF, Rangel A, et al. Prion strain differences in accumulation of PrPSc on neurons and glia are associated with similar expression profiles of neuroinflammatory genes: comparison of three prion strains. PLoS Pathog 2016; 12(4): e1005551.
[62]
Absalon S, Kochanek DM, Raghavan V, et al. MiR-26b, upregulated in Alzheimer’s disease, activates cell cycle entry, tau-phosphorylation, and apoptosis in postmitotic neurons. J Neurosci 2013; 33(37): 14645-59.
[63]
Alam MM, Iqbal S, Naseem I. Ameliorative effect of riboflavin on hyperglycemia, oxidative stress and DNA damage in type-2 diabetic mice: Mechanistic and therapeutic strategies. Arch Biochem Biophys 2015; 584: 10-9.
[64]
Van Everbroeck B, Dewulf E, Pals P, et al. The role of cytokines, astrocytes, microglia and apoptosis in Creutzfeldt-Jakob disease. Neurobiol Aging 2002; 23(1): 59-64.
[65]
Choi SH, Aid S, Caracciolo L, et al. Cyclooxygenase-1 inhibition reduces amyloid pathology and improves memory deficits in a mouse model of Alzheimer’s disease. J Neurochem 2013; 124(1): 59-68.
[66]
Li YY, Cui JG, Hill JM, et al. Increased expression of miRNA-146a in Alzheimer’s disease transgenic mouse models. Neuroscience letters 2011; 487(1): 94-8.
[67]
Gossner AG, Hopkins J. The effect of PrP(Sc) accumulation on inflammatory gene expression within sheep peripheral lymphoid tissue. Vet Microbiol 2015; 181(3-4): 204-11.
[68]
Llorens F, Thune K, Sikorska B, et al. Altered Ca(2+) homeostasis induces Calpain-Cathepsin axis activation in sporadic Creutzfeldt-Jakob disease. Acta Neuropathol Commun 2017; 5(1): 35.
[69]
Forloni G, Angeretti N, Chiesa R, et al. Neurotoxicity of a prion protein fragment. Nature 1993; 362(6420): 543-6.
[70]
Fioriti L, Quaglio E, Massignan T, et al. The neurotoxicity of prion protein (PrP) peptide 106-126 is independent of the expression level of PrP and is not mediated by abnormal PrP species. Mol Cell Neurosci 2005; 28(1): 165-76.
[71]
Ahmad A, Crupi R, Impellizzeri D, et al. Administration of palmitoylethanolamide (PEA) protects the neurovascular unit and reduces secondary injury after traumatic brain injury in mice. Brain Behav Immun 2012; 26(8): 1310-21.
[72]
Sanchez-Rodriguez C, Martin-Sanz E, Cuadrado E, et al. Protective effect of polyphenols on presbycusis via oxidative/nitrosative stress suppression in rats. Exp Gerontol 2016; 83: 31-6.
[73]
Beck AT, Steer RA, Ball R, et al. Comparison of Beck Depression Inventories -IA and -II in psychiatric outpatients. J Pers Assess 1996; 67(3): 588-97.
[74]
Brown DR, Herms JW, Schmidt B, et al. PrP and beta-amyloid fragments activate different neurotoxic mechanisms in cultured mouse cells. Eur J Neurosci 1997; 9(6): 1162-9.
[75]
Christensen KE, Mikael LG, Leung KY, et al. High folic acid consumption leads to pseudo-MTHFR deficiency, altered lipid metabolism, and liver injury in mice. Am J Clin Nutr 2015; 101(3): 646-58.
[76]
Butterfield DA, Hensley K, Harris M, et al. Beta-Amyloid peptide free radical fragments initiate synaptosomal lipoperoxidation in a sequence-specific fashion: Implications to Alzheimer’s disease. Biochem Biophys Res Commun 1994; 200(2): 710-5.
[77]
Butterfield DA, Bush AI. Alzheimer’s amyloid beta-peptide (1-42): Involvement of methionine residue 35 in the oxidative stress and neurotoxicity properties of this peptide. Neurobiol Aging 2004; 25(5): 563-8.
[78]
Varadarajan S, Yatin S, Kanski J, et al. Methionine residue 35 is important in amyloid beta-peptide-associated free radical oxidative stress. Brain Res Bull 1999; 50(2): 133-41.
[79]
Lahiri DK. Prions: A piece of the puzzle? Science 2012; 337(6099): 1172.
[80]
Wong BS, Pan T, Liu T, et al. Prion disease: A loss of antioxidant function? Biochem Biophys Res Commun 2000; 275(2): 249-52.
[81]
Telling GC, Haga T, Torchia M, et al. Interactions between wild-type and mutant prion proteins modulate neurodegeneration in transgenic mice. Genes Dev 1996; 10(14): 1736-50.
[82]
Im N, Zhou W, Na M, et al. Pierisformoside B exhibits neuroprotective and anti-inflammatory effects in murine hippocampal and microglial cells via the HO-1/Nrf2-mediated pathway. Int Immunopharmacol 2014; 24(2): 353-60.
[83]
Jeong JK, Moon MH, Bae BC, et al. Autophagy induced by resveratrol prevents human prion protein-mediated neurotoxicity. Neurosci Res 2012; 73(2): 99-105.
[84]
Cho HJ, Liu G, Jin SM, et al. MicroRNA-205 regulates the expression of Parkinson’s disease-related leucine-rich repeat kinase 2 protein. Hum Mol Genet 2013; 22(3): 608-20.
[85]
Albanes D, Till C, Klein EA, et al. Plasma tocopherols and risk of prostate cancer in the Selenium and Vitamin E Cancer Prevention Trial (SELECT). Cancer Prev Res (Phila) 2014; 7(9): 886-95.
[86]
Ahn JH, Yoo MH, Lee HJ, et al. Coenzyme Q10 in combination with steroid therapy for treatment of sudden sensorineural hearing loss: A controlled prospective study. Clin Otolaryngol 2010; 35(6): 486-9.
[87]
Rambold AS, Miesbauer M, Olschewski D, et al. Green tea extracts interfere with the stress-protective activity of PrP and the formation of PrP. J Neurochem 2008; 107(1): 218-29.
[88]
Shaked GM, Engelstein R, Avraham I, et al. Dimethyl sulfoxide delays PrP sc accumulation and disease symptoms in prion-infected hamsters. Brain Res 2003; 983(1-2): 137-43.
[89]
Prasad KN, Bondy SC. Inhibition of early upstream events in prodromal Alzheimer’s disease by use of targeted antioxidants. Curr Aging Sci 2014; 7(2): 77-90.
[90]
Prasad KN. Simultaneous activation of Nrf2 and elevation of antioxidant compounds for reducing oxidative stress and chronic inflammation in human Alzheimer’s disease. Mech Ageing Dev 2016; 153: 41-7.
[91]
Vile GF, Winterbourn CC. Inhibition of adriamycin-promoted microsomal lipid peroxidation by beta-carotene, alpha-tocopherol and retinol at high and low oxygen partial pressures. FEBS Lett 1988; 238(2): 353-6.
[92]
Niki E. Interaction of ascorbate and alpha-tocopherol. Ann N Y Acad Sci 1987; 498: 186-99.
[93]
Prasad KN. Oxidative stress and pro-inflammatory cytokines may act as one of the signals for regulating microRNAs expression in Alzheimer’s disease. Mech Ageing Dev 2017; 162: 63-71.
[94]
Wu H, Kong L, Tan Y, et al. C66 ameliorates diabetic nephropathy in mice by both upregulating NRF2 function via increase in miR-200a and inhibiting miR-21. Diabetologia 2016; 59(7): 1558-68.
[95]
Jaramillo MC, Zhang DD. The emerging role of the Nrf2-Keap1 signaling pathway in cancer. Genes Dev 2013; 27(20): 2179-91.
[96]
Williamson TP, Johnson DA, Johnson JA. Activation of the Nrf2-ARE pathway by siRNA knockdown of Keap1 reduces oxidative stress and provides partial protection from MPTP-mediated neurotoxicity. Neurotoxicology 2012; 33(3): 272-9.
[97]
Hashimoto M, Yoshimoto M, Sisk A, et al. NACP, a synaptic protein involved in Alzheimer’s disease, is differentially regulated during megakaryocyte differentiation. Biochem Biophys Res Commun 1997; 237(3): 611-6.
[98]
Kirsh VA, Hayes RB, Mayne ST, et al. Supplemental and dietary vitamin E, beta-carotene, and vitamin C intakes and prostate cancer risk. J Natl Cancer Inst 2006; 98(4): 245-54.
[99]
Albini A, Morini M, D’Agostini F, et al. Inhibition of angiogenesis-driven Kaposi’s sarcoma tumor growth in nude mice by oral N-acetylcysteine. Cancer Res 2001; 61(22): 8171-8.
[100]
Ramsey CP, Glass CA, Montgomery MB, et al. Expression of Nrf2 in neurodegenerative diseases. J Neuropathol Exp Neurol 2007; 66(1): 75-85.
[101]
Chen PC, Vargas MR, Pani AK, et al. Nrf2-mediated neuroprotection in the MPTP mouse model of Parkinson’s disease: Critical role for the astrocyte. Proc Natl Acad Sci USA 2009; 106(8): 2933-8.
[102]
Lastres-Becker I, Ulusoy A, Innamorato NG, et al. alpha-Synuclein expression and Nrf2 deficiency cooperate to aggravate protein aggregation, neuronal death and inflammation in early-stage Parkinson’s disease. Hum Mol Genet 2012; 21(14): 3173-92.
[103]
Iwanaga M, Hsu WL, Soda M, et al. Risk of myelodysplastic syndromes in people exposed to ionizing radiation: a retrospective cohort study of Nagasaki atomic bomb survivors. J Clin Oncol 2011; 29(4): 428-34.
[104]
Trujillo J, Chirino YI, Molina-Jijon E, et al. Renoprotective effect of the antioxidant curcumin: Recent findings. Redox Biol 2013; 1: 448-56.
[105]
Steele ML, Fuller S, Patel M, et al. Effect of Nrf2 activators on release of glutathione, cysteinylglycine and homocysteine by human U373 astroglial cells. Redox Biol 2013; 1: 441-5.
[106]
Kode A, Rajendrasozhan S, Caito S, et al. Resveratrol induces glutathione synthesis by activation of Nrf2 and protects against cigarette smoke-mediated oxidative stress in human lung epithelial cells. Am J Physiol Lung Cell Mol Physiol 2008; 294(3): L478-88.
[107]
Gao L, Wang J, Sekhar KR, et al. Novel n-3 fatty acid oxidation products activate Nrf2 by destabilizing the association between Keap1 and Cullin3. J Biol Chem 2007; 282(4): 2529-37.
[108]
Aoi J, Endo M, Kadomatsu T, et al. Angiopoietin-like protein 2 accelerates carcinogenesis by activating chronic inflammation and oxidative stress. Mol Cancer Res 2014; 12(2): 239-49.
[109]
Song J, Kang SM, Lee WT, et al. Glutathione protects brain endothelial cells from hydrogen peroxide-induced oxidative stress by increasing nrf2 expression. Exp Neurobiol 2014; 23(1): 93-103.
[110]
Atkinson J, Kapralov A, Huang Z, et al. Mitochondria-targeted ligands of hene-iron in cytochrome c as novel radioprotectors/radiomitigators. Radiat Res 2010, 56th Annual Meeting Radiation Research Society, Maui, Hawaii, Sept. 25-29 :58a.
[111]
Beriat GK, Ezerarslan H, Akmansu SH, et al. Comparison of efficacy of different treatment methods in the treatment of idiopathic tinnitus. Kulak Burun Bogaz Ihtis Derg 2011; 21(3): 145-53.
[112]
Bai H, Liu R, Chen HL, et al. Enhanced antioxidant effect of caffeic acid phenethyl ester and Trolox in combination against radiation induced-oxidative stress. Chem Biol Interact 2014; 207: 7-15.
[113]
Cui L, Jeong H, Borovecki F, et al. Transcriptional repression of PGC-1alpha by mutant huntingtin leads to mitochondrial dysfunction and neurodegeneration. Cell 2006; 127(1): 59-69.
[114]
Bergstrom P, Andersson HC, Gao Y, et al. Repeated transient sulforaphane stimulation in astrocytes leads to prolonged Nrf2-mediated gene expression and protection from superoxide-induced damage. Neuropharmacology 2011; 60(2-3): 343-53.
[115]
Wruck CJ, Gotz ME, Herdegen T, et al. Kavalactones protect neural cells against amyloid beta peptide-induced neurotoxicity via extracellular signal-regulated kinase 1/2-dependent nuclear factor erythroid 2-related factor 2 activation. Mol Pharmacol 2008; 73(6): 1785-95.
[116]
Kim J, Cha YN, Surh YJ. A protective role of nuclear factor-erythroid 2-related factor-2 (Nrf2) in inflammatory disorders. Mutat Res 2010; 690(1-2): 12-23.
[117]
Li W, Khor TO, Xu C, et al. Activation of Nrf2-antioxidant signaling attenuates NFkappaB-inflammatory response and elicits apoptosis. Biochem Pharmacol 2008; 76(11): 1485-9.
[118]
Abate A, Yang G, Dennery PA, et al. Synergistic inhibition of cyclooxygenase-2 expression by vitamin E and aspirin. Free Radic Biol Med 2000; 29(11): 1135-42.
[119]
Fu Y, Zheng S, Lin J, et al. Curcumin protects the rat liver from CCl4-caused injury and fibrogenesis by attenuating oxidative stress and suppressing inflammation. Mol Pharmacol 2008; 73(2): 399-409.
[120]
Lee HS, Jung KK, Cho JY, et al. Neuroprotective effect of curcumin is mainly mediated by blockade of microglial cell activation. Pharmazie 2007; 62(12): 937-42.
[121]
Rahman S, Bhatia K, Khan AQ, et al. Topically applied vitamin E prevents massive cutaneous inflammatory and oxidative stress responses induced by double application of 12-O-tetradecanoylphorbol-13-acetate (TPA) in mice. Chem Biol Interact 2008; 172(3): 195-205.
[122]
Suzuki YJ, Aggarwal BB, Packer L. Alpha-lipoic acid is a potent inhibitor of NF-kappa B activation in human T cells. Biochem Biophys Res Commun 1992; 189(3): 1709-15.
[123]
Zhu J, Yong W, Wu X, et al. Anti-inflammatory effect of resveratrol on TNF-alpha-induced MCP-1 expression in adipocytes. Biochem Biophys Res Commun 2008; 369(2): 471-7.

© 2024 Bentham Science Publishers | Privacy Policy