Vitagenes, dietary antioxidants and neuroprotection in neurodegenerative diseases(1145 views) Calabrese V, Cornelius C, Mancuso C, Barone E, Calafato S, Bates T, Rizzarelli E, Kostova AT
Frontiers In Bioscience (ISSN: 1093-4715, 1093-9946), 2009 Jan 1; 14: 376-397.
Department of Chemistry, Biochemistry and Mol. Biology Section, Faculty of Medicine, University of Catania, Catania, Italy. calabres@unict.it
Institute of Pharmacology, Catholic University, School of Medicine, Roma, Italy
School of Biomedical Sciences, University of Nottingham, Nottingham NG7 2UH, United Kingdom
Division of Clinical Pharmacology, Department of Medicine and Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD 21205, United States
Biomedical Research Centre, University of Dundee, United Kingdom
References: Mancuso, C., Scapagnini, G., Curro, D., Giuffrida Stella, A.M., De Marco, C., Butterfield, D.A., Calabrese, V., Mitochondrial dysfunction, free radical generation and cellular stress response in neurodegenerative disorders (2007) Frontiers in Bioscience, 12 (3), pp. 1107-1123. , DOI 10.2741/213
Calabrese, V., Scapagnini, G., Colombrita, C., Ravagna, A., Pennisi, G., Giuffrida Stella, A.M., Galli, F., Butterfield, D.A., Redox regulation of heat shock protein expression in aging and neurodegenerative disorders associated with oxidative stress: A nutritional approach (2003) Amino Acids, 25 (3-4), pp. 437-444. , DOI 10.1007/s00726-003-0048-2
Poon, H.F., Calabrese, V., Scapagnini, G., Butterfield, D.A., Free radicals: Key to brain aging and heme oxygenase as a cellular response to oxidative stress (2004) Journals of Gerontology - Series A Biological Sciences and Medical Sciences, 59 (5), pp. 478-493
Forman, H.J., Fukuto, J.M., Torres, M., Redox signaling: Thiol chemistry defines which reactive oxygen and nitrogen species can act as second messengers (2004) American Journal of Physiology - Cell Physiology, 287 (2), pp. C246-C256. , DOI 10.1152/ajpcell.00516.2003
Calabrese, V., Giuffrida Stella, A.M., Butterfield, D.A., Scapagnini, G., Redox regulation in neurodegeneration and longevity: Role of the heme oxygenase and HSP70 systems in brain stress tolerance (2004) Antioxidants and Redox Signaling, 6 (5), pp. 895-913. , DOI 10.1089/ars.2004.6.895
Halliwell, B., Hypothesis: Proteasomal dysfunction. A primary event in neurogeneration that leads to nitrative and oxidative stress and subsequent cell death (2002) Annals of the New York Academy of Sciences, 962, pp. 182-194
Martindale, J.L., Holbrook, N.J., Cellular response to oxidative stress: Signaling for suicide and survival (2002) Journal of Cellular Physiology, 192 (1), pp. 1-15. , DOI 10.1002/jcp.10119
Bergamini, C.M., Gambetti, S., Dondi, A., Cervellati, C., Oxygen, reactive oxygen species and tissue damage (2004) Current Pharmaceutical Design, 10 (14), pp. 1611-1626. , DOI 10.2174/1381612043384664
Pappolla, M.A., Chyan, Y.-J., Omar, R.A., Hsiao, K., Perry, G., Smith, M.A., Bozner, P., Evidence of oxidative stress and in vivo neurotoxicity of β-amyloid in a transgenic mouse model of Alzheimer's disease: A chronic oxidative paradigm for testing antioxidant therapies in vivo (1998) American Journal of Pathology, 152 (4), pp. 871-877
Smith, M.A., Hirai, K., Hsiao, K., Pappolla, M.A., Harris, P.L.R., Siedlak, S.L., Tabaton, M., Perry, G., Amyloid-β deposition in Alzheimer transgenic mice is associated with oxidative stress (1998) Journal of Neurochemistry, 70 (5), pp. 2212-2215
Butterfield, D.A., Drake, J., Pocernich, C., Castegna, A., Evidence of oxidative damage in Alzheimer's disease brain: Central role for amyloid β-peptide (2001) Trends in Molecular Medicine, 7 (12), pp. 548-554. , DOI 10.1016/S1471-4914(01)02173-6, PII S1471491401021736
Butterfield D.Allan, Lauderback, C.M., Lipid peroxidation and protein oxidation in Alzheimer's disease brain: Potential causes and consequences involving amyloid β-peptide-associated free radical oxidative stress (2002) Free Radical Biology and Medicine, 32 (11), pp. 1050-1060. , DOI 10.1016/S0891-5849(02)00794-3, PII S0891584902007943
Mattson, M.P., Pathways towards and away from Alzheimer's disease (2004) Nature, 430 (7000), pp. 631-639. , DOI 10.1038/nature02621
Drew, B., Leeuwenburgh, C., Aging and the role of reactive nitrogen species (2002) Annals of the New York Academy of Sciences, 959, pp. 66-81
Kroncke, K.D., Nitrosative stress and transcription (2003) Biol Chem, 384, pp. 1365-1377
Ridnour, L.A., Thomas, D.D., Mancardi, D., Espey, M.G., Miranda, K.M., Paolocci, N., Feelisch, M., Wink, D.A., The chemistry of nitrosative stress induced by nitric oxide and reactive nitrogen oxide species. Putting perspective on stressful biological situations (2004) Biological Chemistry, 385 (1), pp. 1-10. , DOI 10.1515/BC.2004.001
Calabrese, V., Boyd-Kimball, D., Scapagnini, G., Butterfield, D.A., Nitric oxide and cellular stress response in brain aging and neurodegenerative disorders: The role of vitagenes (2004) In Vivo, 18, pp. 245-267
Calabrese, V., Guagliano, E., Sapienza, M., Panebianco, M., Calafato, S., Puleo, E., Pennisi, G., Stella, A.G., Redox regulation of cellular stress response in aging and neurodegenerative disorders: Role of vitagenes (2007) Neurochemical Research, 32 (4-5), pp. 757-773. , DOI 10.1007/s11064-006-9203-y
Calabrese, V., Butterfield, D.A., Scapagnini, G., Stella, A.M.G., Maines, M.D., Redox regulation of heat shock protein expression by signaling involving nitric oxide and carbon monoxide: Relevance to brain aging, neurodegenerative disorders, and longevity (2006) Antioxidants and Redox Signaling, 8 (3-4), pp. 444-477. , DOI 10.1089/ars.2006.8.444
Mancuso, C., Pani, G., Calabrese, V., Bilirubin: An endogenous scavenger of nitric oxide and reactive nitrogen species (2006) Redox Report, 11 (5), pp. 207-213. , http://www.ingentaconnect.com/content/maney/rer/2006/00000011/00000005/ art00002, DOI 10.1179/135100006X154978
Stocker, R., Antioxidant activities of bile pigments (2004) Antioxidants and Redox Signaling, 6 (5), pp. 841-849. , DOI 10.1089/ars.2004.6.841
Mancuso, C., Heme oxygenase and its products in the nervous system (2004) Antioxidants and Redox Signaling, 6 (5), pp. 878-887. , DOI 10.1089/ars.2004.6.878
Simonian, N.A., Coyle, J.T., Oxidative stress in neurodegenerative diseases (1996) Annual Review of Pharmacology and Toxicology, 36, pp. 83-106
Sayre, L.M., Smith, M.A., Perry, G., Chemistry and biochemistry of oxidative stress in neurodegenerative disease (2001) Current Medicinal Chemistry, 8 (7), pp. 721-738
Andersen, J.K., Oxidative stress in neurodegeneration: Cause or consequence? (2004) Nature Medicine, 10 (SUPPL.), pp. S18-S25
Maines, M.D., (1992) Heme Oxygenase in Clinical Applications and Functions, , Boca Raton: CRC Press
Maines, M.D., The heme oxygenase system: A regulator of second messenger gases (1997) Annual Review of Pharmacology and Toxicology, 37, pp. 517-554
Panahian, N., Yoshiura, M., Maines, M.D., Overexpression of heme oxygenase-1 is neuroprotective in a model of permanent middle cerebral artery occlusion in transgenic mice (1999) Journal of Neurochemistry, 72 (3), pp. 1187-1203
Takeda, A., Perry, G., Abraham, N.G., Dwyer, B.E., Kutty, R.K., Laitinen, J.T., Petersen, R.B., Smith, M.A., Overexpression of heme oxygenase in neuronal cells, the possible interaction with tau (2000) Journal of Biological Chemistry, 275 (8), pp. 5395-5399. , DOI 10.1074/jbc.275.8.5395
Premkumar, D.R., Smith, M.A., Richey, P.L., Petersen, R.B., Castellani, R., Kutty, R.K., Wiggert, B., Kalaria, R.N., Induction of heme oxygenase-1 mRNA and protein in neocortex and cerebral vessels in Alzheimer's disease (1995) J Neurochem, 65, pp. 1399-1402
Schipper, H.M., Heme oxygenase-1: Role in brain aging and neurodegeneration (2000) Exp Gerontol, 35, pp. 821-830
Scapagnini, G., Foresti, R., Calabrese, V., Giuffrida Stella, A.M., Green, C.J., Motterlini, R., Caffeic acid phenethyl ester and curcumin: A novel class of heme oxygenase-1 inducers (2002) Molecular Pharmacology, 61 (3), pp. 554-561. , DOI 10.1124/mol.61.3.554
Mancuso, C., Bonsignore, A., Di Stasio, E., Mordente, A., Motterlini, R., Bilirubin and S-nitrosothiols interaction: Evidence for a possible role of bilirubin as a scavenger of nitric oxide (2003) Biochemical Pharmacology, 66 (12), pp. 2355-2363. , DOI 10.1016/j.bcp.2003.08.022
Ganguli, M., Chandra, V., Kamboh, M.I., Johnston, J.M., Dodge, H.H., Thelma, B.K., Juyal, R.C., DeKosky, S.T., Apolipoprotein E polymorphism and Alzheimer disease: The Indo-US cross- national dementia study (2000) Archives of Neurology, 57 (6), pp. 824-830
Lim, G.P., Chu, T., Yang, F., Beech, W., Frautschy, S.A., Cole, G.M., The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse (2001) Journal of Neuroscience, 21 (21), pp. 8370-8377
Palozza, P., Serini, S., Curro, D., Calviello, G., Igarashi, K., Mancuso, C., β-carotene and cigarette smoke condensate regulate heme oxygenase-1 and its repressor factor Bach1: Relationship with cell growth (2006) Antioxidants and Redox Signaling, 8 (5-6), pp. 1069-1080. , DOI 10.1089/ars.2006.8.1069
Kitamuro, T., Takahashi, K., Ogawa, K., Udono-Fujimori, R., Takeda, K., Furuyama, K., Nakayama, M., Shibahara, S., Bach1 functions as a hypoxia-inducible repressor for the heme oxygenase-1 gene in human cells (2003) Journal of Biological Chemistry, 278 (11), pp. 9125-9133. , DOI 10.1074/jbc.M209939200
Nakayama, M., Takahashi, K., Kitamuro, T., Yasumoto, K.-I., Katayose, D., Shirato, K., Fujii-Kuriyama, Y., Shibahara, S., Repression of heme oxygenase-1 by hypoxia in vascular endothelial cells (2000) Biochemical and Biophysical Research Communications, 271 (3), pp. 665-671. , DOI 10.1006/bbrc.2000.2683
Okinaga, S., Takahashi, K., Takeda, K., Yoshizawa, M., Fujita, H., Sasaki, H., Shibahara, S., Regulation of human heme oxygenase-1 gene expression under thermal stress (1996) Blood, 87 (12), pp. 5074-5084
Takahashi, K., Nakayama, M., Takeda, K., Fujita, H., Shibahara, S., Suppression of heme oxygenase-1 mRNA expression by interferon-γ in human glioblastoma cells (1999) Journal of Neurochemistry, 72 (6), pp. 2356-2361. , DOI 10.1046/j.1471-4159.1999.0722356.x
Oyake, T., Itoh, K., Motohashi, H., Hayashi, N., Hoshino, H., Nishizawa, M., Yamamoto, M., Igarashi, K., Bach proteins belong to a novel family of BTB-basic leucine zipper transcription factors that interact with MafK and regulate transcription through the NF-E2 site (1996) Molecular and Cellular Biology, 16 (11), pp. 6083-6095
Sun, J., Hoshino, H., Takaku, K., Nakajima, O., Muto, A., Suzuki, H., Tashiro, S., Igarashi, K., Hemoprotein Bach1 regulates enhancer availability of heme oxygenase-1 gene (2002) EMBO J, 21, pp. 5216-5224
Shibahara, S., The heme oxygenase dilemma in cellular homeostasis: New insights for the feedback regulation of heme catabolism (2003) Tohoku Journal of Experimental Medicine, 200 (4), pp. 167-186. , DOI 10.1620/tjem.200.167
Wu, L., Wang, R., Carbon monoxide: Endogenous production, physiological functions, and pharmacological applications (2005) Pharmacological Reviews, 57 (4), pp. 585-630. , http://pharmrev.aspetjournals.org/cgi/reprint/57/4/585, DOI 10.1124/pr.57.4.3
Calabrese, V., Mancuso, C., Calvani, M., Rizzarelli, E., Butterfield, D.A., Giuffrida Stella, A.M., Nitric oxide in the central nervous system: Neuroprotection versus neurotoxicity (2007) Nature Reviews Neuroscience, 8 (10), pp. 766-775. , DOI 10.1038/nrn2214, PII NRN2214
Mancuso, C., Perluigi, M., Cini, C., De Marco, C., Stella, A.M.G., Calabrese, V., Heme oxygenase and cyclooxygenase in the central nervous system: A functional interplay (2006) Journal of Neuroscience Research, 84 (7), pp. 1385-1391. , DOI 10.1002/jnr.21049
Kostoglou-Athanassiou, I., Forsling, M.L., Navarra, P., Grossman, A.B., Oxytocin release is inhibited by the generation of carbon monoxide from the rat hypothalamus - Further evidence for carbon monoxide as a neuromodulator (1996) Molecular Brain Research, 42 (2), pp. 301-306. , DOI 10.1016/S0169-328X(96)00137-4, PII S0169328X96001374
Mancuso, C., Kostoglou-Athanassiou, I., Forsling, M.L., Grossman, A.B., Preziosi, P., Navarra, P., Minotti, G., Activation of heme oxygenase and consequent carbon monoxide formation inhibits the release of arginine vasopressin from rat hypothalamic explants. Molecular linkage between heme catabolism and neuroendocrine function (1997) Molecular Brain Research, 50 (1-2), pp. 267-276. , DOI 10.1016/S0169-328X(97)00197-6, PII S0169328X97001976
Mancuso, C., Ragazzoni, E., Tringali, G., Liberale, I., Preziosi, P., Grossman, A., Navarra, P., Inhibition of heme oxygenase in the central nervous system potentiates endotoxin-induced vasopressin release in the rat (1999) Journal of Neuroimmunology, 99 (2), pp. 189-194. , DOI 10.1016/S0165-5728(99)00112-5, PII S0165572899001125
Parkes, D., Kasckow, J., Vale, W., Carbon monoxide modulates secretion of corticotropin-releasing factor from rat hypothalamic cell cultures (1994) Brain Research, 646 (2), pp. 315-318. , DOI 10.1016/0006-8993(94)90097-3
Pozzoli, G., Mancuso, C., Mirtella, A., Preziosi, P., Grossman, A.B., Navarra, P., Carbon monoxide as a novel neuroendocrine modulator: Inhibition of stimulated corticotropin-releasing hormone release from acute rat hypothalamic explants (1994) Endocrinology, 135 (6), pp. 2314-2317. , DOI 10.1210/en.135.6.2314
Mancuso, C., Pistritto, G., Tringali, G., Grossman, A.B., Preziosi, P., Navarra, P., Evidence that carbon monoxide stimulates prostaglandin endoperoxide synthase activity in rat hypothalamic explants and in primary cultures of rat hypothalamic astrocytes (1997) Molecular Brain Research, 45 (2), pp. 294-300. , DOI 10.1016/S0169-328X(96)00258-6, PII S0169328X96002586
Mancuso, C., Tringali, G., Grossman, A., Preziosi, P., Navarra, P., The generation of nitric oxide and carbon monoxide produces opposite effects on the release of immunoreactive interleukin-1β from the rat hypothalamus in vitro: Evidence for the involvement of different signaling pathways (1998) Endocrinology, 139 (3), pp. 1031-1037. , DOI 10.1210/en.139.3.1031
Xi, Q., Tcheranova, D., Parfenova, H., Horowitz, B., Leffler, C.W., Jaggar, J.H., Carbon monoxide activates KCa channels in newborn arteriole smooth muscle cells by increasing apparent Ca2+ sensitivity of α-subunits (2004) American Journal of Physiology - Heart and Circulatory Physiology, 286 (2), pp. H610-H618
Jaggar, J.H., Leffler, C.W., Cheranov, S.Y., Tcheranova, D.E.S., Cheng, X., Carbon monoxide dilates cerebral arterioles by enhancing the coupling of Ca2+ sparks to Ca2+activated K+ channels (2002) Circ Res, 91, pp. 610-617
Otterbein, L.E., Bach, F.H., Alam, J., Soares, M., Lu, H.T., Wysk, M., Davis, R.J., Choi, A.M.K., Carbon monoxide has anti-inflammatory effects involving the mitogen- activated protein kinase pathway (2000) Nature Medicine, 6 (4), pp. 422-428. , DOI 10.1038/74680
Ryter, S.W., Otterbein, L.E., Morse, D., Choi, A.M.K., Heme oxygenase/carbon monoxide signaling pathways: Regulation and functional significance (2002) Molecular and Cellular Biochemistry, 234-235, pp. 249-263. , DOI 10.1023/A:1015957026924
Yenari, M.A., Giffard, R.G., Sapolsky, R.M., Steinberg, G.K., The neuroprotective potential of heat shock protein 70 (HSP70) (1999) Mol Med Today, 5, pp. 525-531
Yenari, M.A., Heat shock proteins and neuroprotection (2002) Advances in Experimental Medicine and Biology, 513, pp. 281-299
Hata, R., Maeda, K., Hermann, D., Mies, G., Hossmann, K.-A., Dynamics of regional brain metabolism and gene expression after middle cerebral artery occlusion in mice (2000) Journal of Cerebral Blood Flow and Metabolism, 20 (2), pp. 306-315
Perez, N., Sugar, J., Charya, S., Johnson, G., Merril, C., Bierer, L., Perl, D., Wallace, W., Increased synthesis and accumulation of heat shock 70 proteins in Alzheimer's disease (1991) Brain Res Mol Brain Res, 1, pp. 249-254
Yoo, B.C., Seidl, R., Cairns, N., Lubec, G., Heat-shock protein 70 levels in brain of patients with Down syndrome and Alzheimer's disease (1999) J Neural Transm Suppl, 57, pp. 315-322
Morrison-Bogorad, M., Zimmerman, A.L., Pardue, S., Heat-shock 70 messenger RNA levels in human brain: Correlation with agonal fever (1995) J Neurochem, 64, pp. 235-246
Kakimura, J.-I., Kitamura, Y., Takata, K., Umeki, M., Suzuki, S., Shibagaki, K., Taniguchi, T., Shimohama, S., Microglial activation and amyloid-β clearance induced by exogenous heat-shock proteins (2002) FASEB Journal, 16 (6), pp. 601-603. , DOI 10.1096/fj.01-0530fje
Calabrese, V., Copani, A., Testa, D., Ravagna, A., Spadaro, F., Tendi, E., Nicoletti, V.G., Giuffrida Stella, A.M., Nitric oxide synthase induction in astroglial cell cultures: Effect on heat shock protein 70 synthesis and oxidant/antioxidant balance (2000) Journal of Neuroscience Research, 60 (5), pp. 613-622. , DOI 10.1002/(SICI)1097-4547(20000601)60:5<613::AID-JNR6>
3.0.CO
Calabrese, V., Testa, G., Ravagna, A., Bates, T.E., Giuffrida Stella, A.M., HSP70 induction in the brain following ethanol administration in the rat: Regulation by glutathione redox state (2000) Biochemical and Biophysical Research Communications, 269 (2), pp. 397-400. , DOI 10.1006/bbrc.2000.2311
Yamawaki, H., Haendeler, J., Berk, B.C., Thioredoxin: A Key Regulator of Cardiovascular Homeostasis (2003) Circulation Research, 93 (11), pp. 1029-1033. , DOI 10.1161/01.RES.0000102869.39150.23
Cho, C.G., Kim, H.J., Chung, S.W., Jung, K.J., Shim, K.H., Yu, B.P., Yodoi, J., Chung, H.Y., Modulation of glutathione and thioredoxin systems by calorie restriction during the aging process (2003) Experimental Gerontology, 38 (5), pp. 539-548. , DOI 10.1016/S0531-5565(03)00005-6
Arner, E.S., Holmgren, A., Physiological functions of thioredoxin and thioredoxin reductase (2000) Eur J Biochem, 267, pp. 6102-6109
Sun, Q.-A., Kirnarsky, L., Sherman, S., Gladyshev, V.N., Selenoprotein oxidoreductase with specificity for thioredoxin and glutathione systems (2001) Proceedings of the National Academy of Sciences of the United States of America, 98 (7), pp. 3673-3678. , DOI 10.1073/pnas.051454398
Bloomfield, K.L., Osborne, S.A., Kennedy, D.D., Clarke, F.M., Tonissen, K.F., Thioredoxin-mediated redox control of the transcription factor Sp1 and regulation of the thioredoxin gene promoter (2003) Gene, 319 (1-2), pp. 107-116. , DOI 10.1016/S0378-1119(03)00799-6
Kim, Y.-C., Yamaguchi, Y., Kondo, N., Masutani, H., Yodoi, J., Thioredoxin-dependent redox regulation of the antioxidant responsive element (ARE) in electrophile response (2003) Oncogene, 22 (12), pp. 1860-1865. , DOI 10.1038/sj.onc.1206369
Tanito, M., Masutani, H., Kim, Y.-C., Nishikawa, M., Ohira, A., Yodoi, J., Sulforaphane induces thioredoxin through the antioxidant-responsive element and attenuates retinal light damage in mice (2005) Investigative Ophthalmology and Visual Science, 46 (3), pp. 979-987. , DOI 10.1167/iovs.04-1120
Eftekharpour, E., Holmgren, A., Juurlink, B.H., Thioredoxin reductase and glutathione synthesis is upregulated by t-butylhydroquinone in cortical astrocytes but not in cortical neurons (2000) Glia, 31, pp. 241-248
Hintze, K.J., Wald, K.A., Zeng, H., Jeffery, E.H., Finley, J.W., Thioredoxin reductase in human hepatoma cells is transcriptionally regulated by sulforaphane and other electrophiles via an antioxidant response element (2003) Journal of Nutrition, 133 (9), pp. 2721-2727
Dinkova-Kostova, A.T., Cheah, J., Samouilov, A., Zweier, J.L., Bozak, R.E., Hicks, R.J., Talalay, P., Phenolic Michael reaction acceptors: Combined direct and indirect antioxidant defenses against electrophiles and oxidants (2007) Medicinal Chemistry, 3 (3), pp. 261-268. , http://www.ingentaconnect.com/content/ben/mc/2007/00000003/00000003/ art00006, DOI 10.2174/157340607780620680
Sakurai, A., Nishimoto, M., Himeno, S., Imura, N., Tsujimoto, M., Kunimoto, M., Hara, S., Transcriptional regulation of thioredoxin reductase 1 expression by cadmium in vascular endothelial cells: Role of NF-E2-related factor-2 (2005) Journal of Cellular Physiology, 203 (3), pp. 529-537. , DOI 10.1002/jcp.20246
Hirota, K., Nakamura, H., Masutani, H., Yodoi, J., Thioredoxin superfamily and thioredoxin-inducing agents (2002) Annals of the New York Academy of Sciences, 957, pp. 189-199
Baker, A.F., Dragovich, T., Tate, W.R., Ramanathan, R.K., Roe, D., Hsu, C.-H., Kirkpatrick, D.L., Powis, G., The antitumor thioredoxin-1 inhibitor PX-12 (1-methylpropyl 2-imidazolyl disulfide) decreases thioredoxin-1 and VEGF levels in cancer patient plasma (2006) Journal of Laboratory and Clinical Medicine, 147 (2), pp. 83-90. , DOI 10.1016/j.lab.2005.09.001, PII S0022214305003100
Nakamura, H., Bai, J., Nishinaka, Y., Ueda, S., Sasada, T., Ohshio, G., Imamura, M., Yodoi, J., Expression of thioredoxin and glutaredoxin, redox-regulating proteins, in pancreatic cancer (2000) Cancer Detection and Prevention, 24 (1), pp. 53-60
Haapasalo, H., Kylaniemi, M., Paunu, N., Kinnula, V.L., Soini, Y., Expression of antioxidant enzymes in astrocytic brain tumors (2003) Brain Pathology, 13 (2), pp. 155-164
Berggren, M.M., Powis, G., Alternative splicing is associated with decreased expression of the redox proto-oncogene thioredoxin-1 in human cancers (2001) Archives of Biochemistry and Biophysics, 389 (1), pp. 144-149. , DOI 10.1006/abbi.2001.2325
Biaglow, J.E., Miller, R.A., The thioredoxin reductase/thioredoxin system: Novel redox targets for cancer therapy (2005) Cancer Biology and Therapy, 4 (1), pp. 6-13. , http://www.landesbioscience.com/journals/cbt/cbtpdf/ a11820916767671121y00gf30471010/biaglow4-1.pdf
Bai, J., Nakamura, H., Kwon, Y.-W., Hattori, I., Yamaguchi, Y., Kim, Y.-C., Kondo, N., Yodoi, J., Critical roles of thioredoxin in nerve growth factor-mediated signal transduction and neurite outgrowth in PC12 cells (2003) Journal of Neuroscience, 23 (2), pp. 503-509
Masutani, H., Bai, J., Kim, Y.-C., Yodoi, J., Thioredoxin as a neurotrophic cofactor and an important regulator of neuroprotection (2004) Molecular Neurobiology, 29 (3), pp. 229-242. , DOI 10.1385/MN:29:3:229
Trigona, W.L., Mullarky, I.K., Cao, Y., Sordillo, L.M., Thioredoxin reductase regulates the induction of haem oxygenase-1 expression in aortic endothelial cells (2006) Biochemical Journal, 394 (1), pp. 207-216. , DOI 10.1042/BJ20050712
Satoh, T., Ishige, K., Sagara, Y., Protective effects on neuronal cells of mouse afforded by ebselen against oxidative stress at multiple steps (2004) Neuroscience Letters, 371 (1), pp. 1-5. , DOI 10.1016/j.neulet.2004.04.055, PII S0304394004005063
Das, K.C., Das, C.K., Thioredoxin, a singlet oxygen quencher and hydroxyl radical scavenger: Redox independent functions (2000) Biochem Biophys Res Commun, 277, pp. 443-447
Ju, T.C., Chen, S.D., Liu, C.C., Yang, D.I., Protective effects of S-nitrosoglutathione against amyloid beta-peptide neurotoxicity (2005) Free Radic Biol Med, 38, pp. 938-949
Rauhala, P., Andoh, T., Yeh, K., Chiueh, C.C., Contradictory effects of sodium nitroprusside and S-nitroso-N- acetylpenicillamine on oxidative stress in brain dopamine neurons in vivo (2002) Annals of the New York Academy of Sciences, 962, pp. 60-72
Lee, S.Y., Andoh, T., Murphy, D.L., Chiueh, C.C., 17betaestradiol activates ICI 182, 780-sensitive estrogen receptors and cyclic GMP-dependent thioredoxin expression for neuroprotection (2003) FASEB J, 17, pp. 947-948
Rahman, I., Biswas, S.K., Kirkham, P.A., Regulation of inflammation and redox signaling by dietary polyphenols (2006) Biochemical Pharmacology, 72 (11), pp. 1439-1452. , DOI 10.1016/j.bcp.2006.07.004, PII S0006295206004199, Cell Signalling, Transcription and Translation as Therapeutic Tergets
Sharma, R.A., Gescher, A.J., Steward, W.P., Curcumin: The story so far (2005) European Journal of Cancer, 41 (13), pp. 1955-1968. , DOI 10.1016/j.ejca.2005.05.009
Ravindranath, V., Chandrasekhara, N., Metabolism of curcumin-studies with [3H]curcumin (1981) Toxicology, 22, pp. 337-344
Maiti, K., Mukherjee, K., Gantait, A., Saha, B.P., Mukherjee, P.K., Curcumin-phospholipid complex: Preparation, therapeutic evaluation and pharmacokinetic study in rats (2007) International Journal of Pharmaceutics, 330 (1-2), pp. 155-163. , DOI 10.1016/j.ijpharm.2006.09.025, PII S0378517306007411
Shoba, G., Joy, D., Joseph, T., Majeed, M., Rajendran, R., Srinivas, P.S.S.R., Influence of piperine on the pharmacokinetics of curcumin in animals and human volunteers (1998) Planta Medica, 64 (4), pp. 353-356. , DOI 10.1055/s-2006-957450
Cheng, A.L., Hsu, C.H., Lin, J.K., Hsu, M.M., Ho, Y.F., Shen, T.S., Ko, J.Y., Hsieh, C.Y., Phase I clinical trial of curcumin, a chemopreventive agent, in patients with high-risk or pre-malignant lesions (2001) Anticancer Res, 21, pp. 2895-2900
Garcia-Alloza, M., Borrelli, L.A., Rozkalne, A., Hyman, B.T., Bacskai, B.J., Curcumin labels amyloid pathology in vivo, disrupts existing plaques, and partially restores distorted neurites in an Alzheimer mouse model (2007) Journal of Neurochemistry, 102 (4), pp. 1095-1104. , DOI 10.1111/j.1471-4159.2007.04613.x
Somparn, P., Phisalaphong, C., Nakornchai, S., Unchern, S., Morales, N.P., Comparative antioxidant activities of curcumin and its demethoxy and hydrogenated derivatives (2007) Biological and Pharmaceutical Bulletin, 30 (1), pp. 74-78. , http://www.jstage.jst.go.jp/article/bpb/30/1/74/_pdf, DOI 10.1248/bpb.30.74
Ireson, C.R., Jones, D.J.L., Orr, S., Coughtrie, M.W.H., Boocock, D.J., Williams, M.L., Farmer, P.B., Gescher, A.J., Metabolism of the cancer chemopreventive agent curcumin in human and rat intestine (2002) Cancer Epidemiology Biomarkers and Prevention, 11 (1), pp. 105-111
Lin, J.K., Pan, M.H., Lin-Shiau, S.Y., Recent studies on the biofunctions and biotransformations of curcumin (2000) Biofactors, 13, pp. 153-158
Hayeshi, R., Mutingwende, I., Mavengere, W., Masiyanise, V., Mukanganyama, S., The inhibition of human glutathione S-transferases activity by plant polyphenolic compounds ellagic acid and curcumin (2007) Food and Chemical Toxicology, 45 (2), pp. 286-295. , DOI 10.1016/j.fct.2006.07.027, PII S0278691506002407
Thapliyal, R., Maru, G.B., Inhibition of cytochrome P450 isozymes by curcumins in vitro and in vivo (2001) Food and Chemical Toxicology, 39 (6), pp. 541-547. , DOI 10.1016/S0278-6915(00)00165-4, PII S0278691500001654
Basu, N.K., Ciotti, M., Hwang, M.S., Kole, L., Mitra, P.S., Cho, J.W., Owens, I.S., Differential and Special Properties of the Major Human UGT1-encoded Gastrointestinal UDP-glucuronosyltransferases Enhance Potential to Control Chemical Uptake (2004) Journal of Biological Chemistry, 279 (2), pp. 1429-1441. , DOI 10.1074/jbc.M306439200
Oetari, S., Sudibyo, M., Commandeur, J.N.M., Samhoedi, R., Vermeulen, N.P.E., Effects of curcumin on cytochrome P450 and glutathione S-transferase activities in rat liver (1996) Biochemical Pharmacology, 51 (1), pp. 39-45. , DOI 10.1016/0006-2952(95)02113-2
Sharma, R.A., Euden, S.A., Platton, S.L., Cooke, D.N., Shafayat, A., Hewitt, H.R., Marczylo, T.H., Steward, W.P., Phase I clinical trial of oral curcumin: Biomarkers of systemic activity and compliance (2004) Clinical Cancer Research, 10 (20), pp. 6847-6854. , DOI 10.1158/1078-0432.CCR-04-0744
Deodhar, S.D., Sethi, R., Srimal, R.C., Preliminary study on antirheumatic activity of curcumin (diferuloyl methane) (1980) Indian Journal of Medical Research, 71 (4), pp. 632-634
Sugiyama, Y., Kawakishi, S., Osawa, T., Involvement of the β-diketone moiety in the antioxidative mechanism of tetrahydrocurcumin (1996) Biochemical Pharmacology, 52 (4), pp. 519-525. , DOI 10.1016/0006-2952(96)00302-4
Osawa, T., Sugiyama, Y., Inayoshi, M., Kawakishi, S., Antioxidative activity of tetrahydrocurcuminoids (1995) Biosci Biotechnol Biochem, 59, pp. 1609-1612
Chen, W.-F., Deng, S.-L., Zhou, B., Yang, L., Liu, Z.-L., Curcumin and its analogues as potent inhibitors of low density lipoprotein oxidation: H-atom abstraction from the phenolic groups and possible involvement of the 4-hydroxy-3-methoxyphenyl groups (2006) Free Radical Biology and Medicine, 40 (3), pp. 526-535. , DOI 10.1016/j.freeradbiomed.2005.09.008, PII S0891584905005265
Priyadarsini, K.I., Maity, D.K., Naik, G.H., Kumar, M.S., Unnikrishnan, M.K., Satav, J.G., Mohan, H., Role of phenolic O-H and methylene hydrogen on the free radical reactions and antioxidant activity of curcumin (2003) Free Radical Biology and Medicine, 35 (5), pp. 475-484. , DOI 10.1016/S0891-5849(03)00325-3
Reyes-Gordillo, K., Segovia, J., Shibayama, M., Vergara, P., Moreno, M.G., Muriel, P., Curcumin protects against acute liver damage in the rat by inhibiting NF-κB, proinflammatory cytokines production and oxidative stress (2007) Biochimica et Biophysica Acta - General Subjects, 1770 (6), pp. 989-996. , DOI 10.1016/j.bbagen.2007.02.004, PII S0304416507000529
Venkataranganna, M.V., Rafiq, Md., Gopumadhavan, S., Peer, G., Babu, U.V., Mitra, S.K., NCB-02 (standardized Curcumin preparation) protects dinitrocholorobenzene-induced colitis through down-regulation of NFκ-B and iNOS (2007) World Journal of Gastroenterology, 13 (7), pp. 1103-1107
Shishodia, S., Potdar, P., Gairola, C.G., Aggarwal, B.B., Curcumin (diferuloylmethane) down-regulates cigarette smoke-induced NF-κB activation through inhibition of IκBα kinase in human lung epithelial cells: Correlation with suppression of COX-2, MMP-9 and cyclin D1 (2003) Carcinogenesis, 24 (7), pp. 1269-1279. , DOI 10.1093/carcin/bgg078
Bhattacharyya, S., Mandal, D., Sen, G.S., Pal, S., Banerjee, S., Lahiry, L., Finke, J.H., Sa, G., Tumor-induced oxidative stress perturbs nuclear factor-κB activity-augmenting tumor necrosis factor-α-mediated T-cell death: Protection by curcumin (2007) Cancer Research, 67 (1), pp. 362-370. , DOI 10.1158/0008-5472.CAN-06-2583
Choi, H., Chun, Y.S., Kim, S.W., Kim, M.S., Park, J.W., Curcumin inhibits hypoxia-inducible factor-1 by degrading aryl hydrocarbon receptor nuclear translocator: A mechanism of tumor growth inhibition (2006) Mol Pharmacol, 70, pp. 1664-1671
Abuarqoub, H., Green, C.J., Foresti, R., Motterlini, R., Curcumin reduces cold storage-induced damage in human cardiac myoblasts (2007) Experimental and Molecular Medicine, 39 (2), pp. 139-148. , http://www.e-emm.org/article/article_files/2.%20139-148.pdf
Jeong, G.-S., Oh, G.-S., Pae, H.-O., Jeong, S.-O., Kim, Y.-C., Shin, M.-K., Seo, B.Y., Chung, H.-T., Comparative effects of curcuminoids on endothelial heme oxygenase-1 expression: Ortho-methoxy groups are essential to enhance heme oxygenase activity and protection (2006) Journal of the Chinese Medical Association, 38 (4), pp. 393-400
McNally, S.J., Harrison, E.M., Ross, J.A., Garden, O.J., Wigmore, S.J., Curcumin induces heme oxygenase-1 in hepatocytes and is protective in simulated cold preservation and warm reperfusion injury (2006) Transplantation, 81, pp. 623-626
Rushworth, S.A., Ogborne, R.M., Charalambos, C.A., O'Connell, M.A., Role of protein kinase C δ in curcumin-induced antioxidant response element-mediated gene expression in human monocytes (2006) Biochemical and Biophysical Research Communications, 341 (4), pp. 1007-1016. , DOI 10.1016/j.bbrc.2006.01.065, PII S0006291X0600146X
Balogun, E., Foresti, R., Green, C.J., Motterlini, R., Changes in temperature modulate heme oxygenase-1 induction by curcumin in renal epithelial cells (2003) Biochemical and Biophysical Research Communications, 308 (4), pp. 950-955. , DOI 10.1016/S0006-291X(03)01517-1
Andreadi, C.K., Howells, L.M., Atherfold, P.A., Manson, M.M., Involvement of Nrf2, p38, B-Raf, and nuclear factor-κB, but not phosphatidylinositol 3-kinase, in induction of hemeoxygenase-1 by dietary polyphenols (2006) Molecular Pharmacology, 69 (3), pp. 1033-1040. , http://molpharm.aspetjournals.org/cgi/reprint/69/3/1033
Rashmi, R., Santhosh Kumar, T.R., Karunagaran, D., Human colon cancer cells differ in their sensitivity to curcumin-induced apoptosis and heat shock protects them by inhibiting the release of apoptosis-inducing factor and caspases (2003) FEBS Letters, 538 (1-3), pp. 19-24. , DOI 10.1016/S0014-5793(03)00099-1
Sood, A., Mathew, R., Trachtman, H., Cytoprotective effect of curcumin in human proximal tubule epithelial cells exposed to Shiga toxin (2001) Biochemical and Biophysical Research Communications, 283 (1), pp. 36-41. , DOI 10.1006/bbrc.2001.4749
Chen, Y.C., Tsai, S.H., Shen, S.C., Lin, J.K., Lee, W.R., Alternative activation of extracellular signal-regulated protein kinases in curcumin and arsenite-induced HSP70 gene expression in human colorectal carcinoma cells (2001) Eur J Cell Biol, 80, pp. 213-221
Chen, Y.C., Kuo, T.C., Lin-Shiau, S.Y., Lin, J.K., Induction of HSP70 gene expression by modulation of Ca (+2) ion and cellular p53 protein by curcumin in colorectal carcinoma cells (1996) Mol Carcinog, 17, pp. 224-234
Kato, K., Ito, H., Kamei, K., Iwamoto, I., Stimulation of the stress-induced expression of stress proteins by curcumin in cultured cells and in rat tissues in vivo (1998) Cell Stress and Chaperones, 3 (3), pp. 152-160. , DOI 10.1379/1466-1268(1998)003<0152:SOTSIE>
2.3.CO
Fang, J., Lu, J., Holmgren, A., Thioredoxin reductase is irreversibly modified by curcumin: A novel molecular mechanism for its anticancer activity (2005) Journal of Biological Chemistry, 280 (26), pp. 25284-25290. , DOI 10.1074/jbc.M414645200
Nguyen, T., Sherratt, P.J., Pickett, C.B., Regulatory Mechanisms Controlling Gene Expression Mediated by the Antioxidant Response Element (2003) Annual Review of Pharmacology and Toxicology, 43, pp. 233-260. , DOI 10.1146/annurev.pharmtox.43.100901.140229
Nioi, P., McMahon, M., Itoh, K., Yamamoto, M., Hayes, J.D., Identification of a novel NRF2-regulated antioxidant response element (ARE) in the mouse NAD(P)H:quinone oxidoreductase 1 gene: Reassessment of the ARE consensus sequence (2003) Biochemical Journal, 374 (2), pp. 337-348. , DOI 10.1042/BJ20030754
Prestera, T., Talalay, P., Alam, J., Ahn, Y.I., Lee, P.J., Choi, A.M., Parallel induction of heme oxygenase-1 and chemoprotective phase 2 enzymes by electrophiles and antioxidants: Regulation by upstream antioxidant-responsive elements (ARE) (1995) Molecular Medicine, 1 (7), pp. 827-837
Moi, P., Chan, K., Asunis, I., Cao, A., Kan, Y.W., Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the β-globin locus control region (1994) Proceedings of the National Academy of Sciences of the United States of America, 91 (21), pp. 9926-9930. , DOI 10.1073/pnas.91.21.9926
Itoh, K., Wakabayashi, N., Katoh, Y., Ishii, T., Igarashi, K., Engel, J.D., Yamamoto, M., Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain (1999) Genes and Development, 13 (1), pp. 76-86
Motohashi, H., Yamamoto, M., Nrf2-Keap1 defines a physiologically important stress response mechanism (2004) Trends in Molecular Medicine, 10 (11), pp. 549-557. , DOI 10.1016/j.molmed.2004.09.003, PII S1471491404002357
Katsuoka, F., Motohashi, H., Ishii, T., Aburatani, H., Engel, J.D., Yamamoto, M., Genetic evidence that small Maf proteins are essential for the activation of antioxidant response element-dependent genes (2005) Molecular and Cellular Biology, 25 (18), pp. 8044-8051. , DOI 10.1128/MCB.25.18.8044-8051.2005
Lee, J.-M., Li, J., Johnson, D.A., Stein, T.D., Kraft, A.D., Calkins, M.J., Jakel, R.J., Johnson, J.A., Nrf2, a multi-organ protector? (2005) FASEB Journal, 19 (9), pp. 1061-1066. , DOI 10.1096/fj.04-2591hyp
Wakabayashi, N., Dinkova-Kostova, A.T., Holtzclaw, W.D., Kang, M.-I., Kobayashi, A., Yamamoto, M., Kensler, T.W., Talalay, P., Protection against electrophile and oxidant stress by induction of the phase 2 response: Fate of cysteines of the Keap1 sensor modified by inducers (2004) Proceedings of the National Academy of Sciences of the United States of America, 101 (7), pp. 2040-2045. , DOI 10.1073/pnas.0307301101
Kensler, T.W., Wakabayashi, N., Biswal, S., Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway (2007) Annu Rev Pharmacol Toxicol, 47, pp. 89-116
Rangasamy, T., Cho, C.Y., Thimmulappa, R.K., Zhen, L., Srisuma, S.S., Kensler, T.W., Yamamoto, M., Biswal, S., Genetic ablation of Nrf2 enhances susceptibility to cigarette smoke-induced emphysema in mice (2004) Journal of Clinical Investigation, 114 (9), pp. 1248-1259. , DOI 10.1172/JCI200421146
Rangasamy, T., Guo, J., Mitzner, W.A., Roman, J., Singh, A., Fryer, A.D., Yamamoto, M., Biswal, S., Disruption of Nrf2 enhances susceptibility to severe airway inflammation and asthma in mice (2005) Journal of Experimental Medicine, 202 (1), pp. 47-59. , http://www.jem.org/cgi/reprint/202/1/47.pdf, DOI 10.1084/jem.20050538
Thimmulappa, R.K., Lee, H., Rangasamy, T., Reddy, S.P., Yamamoto, M., Kensler, T.W., Biswal, S., Nrf2 is a critical regulator of the innate immune response and survival during experimental sepsis (2006) J Clin Invest, 116, pp. 984-995
Kobayashi, M., Yamamoto, M., Nrf2-Keap1 regulation of cellular defense mechanisms against electrophiles and reactive oxygen species (2006) Advances in Enzyme Regulation, 46 (1), pp. 113-140. , DOI 10.1016/j.advenzreg.2006.01.007, PII S0065257106000094, Proceedings of teh 46th International Sumposium on Regulation of the Enzym Activity ans Synthesis in Normal and Neoplastic Tissues
Zhang, X., Lu, L., Dixon, C., Wilmer, W., Song, H., Chen, X., Rovin, B.H., Stress protein activation by the cyclopentenone prostaglandin 15-deoxy-Δ12,14-prostaglandin J2 in human mesangial cells (2004) Kidney International, 65 (3), pp. 798-810. , DOI 10.1111/j.1523-1755.2004.00454.x
Rokutan, K., Miyoshi, M., Teshima, S., Kawai, T., Kawahara, T., Kishi, K., Phenylarsine oxide inhibits heat shock protein 70 induction in cultured guinea pig gastric mucosal cells (2000) Am J Physiol Cell Physiol, 279, pp. C1506-C1515
Liu, H., Lightfoot, R., Stevens, J.L., Activation of heat shock factor by alkylating agents is triggered by glutathione depletion and oxidation of protein thiols (1996) Journal of Biological Chemistry, 271 (9), pp. 4805-4812. , DOI 10.1074/jbc.271.9.4805
Tabner, B.J., Turnbull, S., El-Agnaf, O., Allsop, D., Production of reactive oxygen species from aggregating proteins implicated in Alzheimer's disease, Parkinson's disease and other neurodegenerative diseases (2001) Curr Top Med Chem, 1, pp. 507-517
Barnham, K.J., Cappai, R., Beyreuther, K., Masters, C.L., Hill, A.F., Delineating common molecular mechanisms in Alzheimer's and prion diseases (2006) Trends in Biochemical Sciences, 31 (8), pp. 465-472. , DOI 10.1016/j.tibs.2006.06.006, PII S096800040600168X
Hinault, M.-P., Ben-Zvi, A., Goloubinoff, P., Chaperones and proteases: Cellular fold-controlling factors of proteins in neurodegenerative diseases and aging (2006) Journal of Molecular Neuroscience, 30 (3), pp. 249-265. , DOI 10.1385/JMN:30:3:249, PII JMN303249
Zhang, K., Kaufman, R.J., The unfolded protein response: A stress signaling pathway critical for health and disease (2006) Neurology, 66 (2 SUPPL. 1), pp. S102-S109. , PII 0000611420060124100018
Nakamura, T., Lipton, S.A., Molecular mechanisms of nitrosative stress-mediated protein misfolding in neurodegenerative diseases (2007) Cellular and Molecular Life Sciences, 64 (13), pp. 1609-1620. , DOI 10.1007/s00018-007-6525-0
Schroder, M., The unfolded protein response (2006) Molecular Biotechnology, 34 (2), pp. 279-290. , DOI 10.1385/MB:34:2:279, PII MB342279
Schroder, M., Kaufman, R.J., The mammalian unfolded protein response (2005) Annual Review of Biochemistry, 74, pp. 739-789. , DOI 10.1146/annurev.biochem.73.011303.074134
Calabrese, V., Sultana, R., Scapagnini, G., Guagliano, E., Sapienza, M., Bella, R., Kanski, J., Butterfield, D.A., Nitrosative stress, cellular stress response, and thiol homeostasis in patients with Alzheimer's disease (2006) Antioxidants and Redox Signaling, 8 (11-12), pp. 1975-1986
Katzman, R., Saitoh, T., Advances in Alzheimer's disease (1991) FASEB Journal, 5 (3), pp. 278-286
Guix, F.X., Uribesalgo, I., Coma, M., Munoz, F.J., The physiology and pathophysiology of nitric oxide in the brain (2005) Progress in Neurobiology, 76 (2), pp. 126-152. , DOI 10.1016/j.pneurobio.2005.06.001, PII S030100820500064X
Kim, D.S.H.L., Park, S.-Y., Kim, J.-Y., Curcuminoids from Curcuma longa L. (Zingiberaceae) that protect PC12 rat pheochromocytoma and normal human umbilical vein endothelial cells from βA(1-42) insult (2001) Neuroscience Letters, 303 (1), pp. 57-61. , DOI 10.1016/S0304-3940(01)01677-9, PII S0304394001016779
Ali, R.E., Rattan, S.I.S., Curcumin's biphasic hormetic response on proteasome activity and heat-shock protein synthesis in human keratinocytes (2006) Annals of the New York Academy of Sciences, 1067 (1), pp. 394-399. , DOI 10.1196/annals.1354.056
Yang, F., Lim, G.P., Begum, A.N., Ubeda, O.J., Simmons, M.R., Ambegaokar, S.S., Chen, P., Cole, G.M., Curcumin inhibits formation of amyloid β oligomers and fibrils, binds plaques, and reduces amyloid in vivo (2005) Journal of Biological Chemistry, 280 (7), pp. 5892-5901. , DOI 10.1074/jbc.M404751200
Voelker, R., Parkinson disease guidelines aid diagnosis, management (2006) JAMA, 295, pp. 2126-2128
Przedborski, S., Ischiropoulos, H., Reactive oxygen and nitrogen species: Weapons of neuronal destruction in models of Parkinson's disease (2005) Antioxidants and Redox Signaling, 7 (5-6), pp. 685-693. , DOI 10.1089/ars.2005.7.685
Hald, A., Lotharius, J., Oxidative stress and inflammation in Parkinson's disease: Is there a causal link? (2005) Experimental Neurology, 193 (2), pp. 279-290. , DOI 10.1016/j.expneurol.2005.01.013
Burns, R.S., Chiueh, C.C., Markey, S.P., A primate model of parkinsonism: Selective destruction of dopaminergic neurons in the pars compacta of the substantia nigra by N-methyl-4-phenyl-1,2,3, 6-tetrahydropyridine (1983) Proceedings of the National Academy of Sciences of the United States of America, 80 (I14), pp. 4546-4550
Chiueh, C.C., Andoh, T., Lai, A.R., Lai, E., Krishna, G., Neuroprotective strategies in Parkinson's disease: Protection against progressive nigral damage induced by free radicals (2000) Neurotox Res, 2, pp. 293-310
Rajeswari, A., Curcumin protects mouse brain from oxidative stress caused by 1-methyl-4-phenyl-1,2,3,6-tetrahydro pyridine (2006) European Review for Medical and Pharmacological Sciences, 10 (4), pp. 157-161
Chen, J., Tang, X.Q., Zhi, J.L., Cui, Y., Yu, H.M., Tang, E.H., Sun, S.N., Chen, P.X., Curcumin protects PC12 cells against 1-methyl-4-phenylpyridinium ion-induced apoptosis by bcl-2-mitochondria-ROS-iNOS pathway (2006) Apoptosis, 11 (6), pp. 943-953. , DOI 10.1007/s10495-006-6715-5
Mythri, R.B., Jagatha, B., Pradhan, N., Andersen, J., Bharath, M.M.S., Mitochondrial complex I inhibition in Parkinson's disease: How can curcumin protect mitochondria? (2007) Antioxidants and Redox Signaling, 9 (3), pp. 399-408. , DOI 10.1089/ars.2006.1479
Al-Omar, F.A., Nagi, M.N., Abdulgadir, M.M., Al Joni, K.S., Al-Majed, A.A., Immediate and delayed treatments with curcumin prevents forebrain ischemia-induced neuronal damage and oxidative insult in the rat hippocampus (2006) Neurochemical Research, 31 (5), pp. 611-618. , DOI 10.1007/s11064-006-9059-1
Wang, Q., Sun, A.Y., Simonyi, A., Jensen, M.D., Shelat, P.B., Rottinghaus, G.E., MacDonald, R.S., Sun, G.Y., Neuroprotective mechanisms of curcumin against cerebral ischemia-induced neuronal apoptosis and behavioral deficits (2005) Journal of Neuroscience Research, 82 (1), pp. 138-148. , DOI 10.1002/jnr.20610
Ghoneim, A.I., Abdel-Naim, A.B., Khalifa, A.E., El-Denshary, E.S., Protective effects of curcumin against ischaemia/reperfusion insult in rat forebrain (2002) Pharmacological Research, 46 (3), pp. 273-279. , DOI 10.1016/S1043-6618(02)00123-8
Evans, A.M., Fornasini, G., Pharmacokinetics of L-carnitine (2003) Clinical Pharmacokinetics, 42 (11), pp. 941-967. , DOI 10.2165/00003088-200342110-00002
Rebouche, C.J., Kinetics, pharmacokinetics, and regulation of L-Carnitine and acetyl-L-carnitine metabolism (2004) Annals of the New York Academy of Sciences, 1033, pp. 30-41. , DOI 10.1196/annals.1320.003
Lombard, K.A., Olson, L., Nelson, S.E., Rebouche, C.J., Carnitine status of lactoovovegetarians and strict vegetarian adults and children (1989) American Journal of Clinical Nutrition, 50 (2), pp. 301-306
Brass, E.P., Hoppel, C.L., Hiatt, W.R., Effect of intravenous L-carnitine on carnitine homeostasis and fuel metabolism during exercise in humans (1994) Clinical Pharmacology and Therapeutics, 55 (6), pp. 681-692
Gross, C.J., Savaiano, D.A., Effect of development and nutritional state on the uptake metabolism and release of free and acetyl-L-carnitine by the rodent small intestine (1993) Biochimica et Biophysica Acta - Lipids and Lipid Metabolism, 1170 (3), pp. 265-274. , DOI 10.1016/0005-2760(93)90009-X
Parnetti, L., Gaiti, A., Mecocci, P., Cadini, D., Senin, U., Pharmacokinetics of IV and oral acetyl-L-carnitine in a multiple dose regimen in patients with senile dementia of Alzheimer type (1992) Eur J Clin Pharmacol, 42, pp. 89-93
Kelly, J.G., Hunt, S., Doyle, G.D., Laher, M.S., Carmody, M., Marzo, A., Arrigoni Martelli, E., Pharmacokinetics of oral acetyl-L-carnitine in renal impairment (1990) European Journal of Clinical Pharmacology, 38 (3), pp. 309-312
McDaniel, M.A., Maier, S.F., Einstein, G.O., "Brainspecific" nutrients: A memory cure? (2003) Nutrition, 19, pp. 957-975
Abdul, H.M., Calabrese, V., Calvani, M., Butterfield, D.A., Acetyl-L-carnitine-induced up-regulation of heat shock proteins protects cortical neurons against amyloid-beta peptide 1-42-mediated oxidative stress and neurotoxicity: Implications for Alzheimer's disease (2006) Journal of Neuroscience Research, 84 (2), pp. 398-408. , DOI 10.1002/jnr.20877
Calabrese, V., Colombrita, C., Sultana, R., Scapagnini, G., Calvani, M., Butterfield, D.A., Giuffrida Stella, A.M., Redox modulation of heat shock protein expression by acetylcarnitine in aging brain: Relationship to antioxidant status and mitochondrial function (2006) Antioxidants and Redox Signaling, 8 (3-4), pp. 404-416. , DOI 10.1089/ars.2006.8.404
Rai, G., Wright, G., Scott, L., Beston, B., Rest, J., Exton-Smith, A.N., Double-blind, placebo controlled study of acetyl-l-carnitine in patients with Alzheimer's dementia (1990) Curr Med Res Opin, 11, pp. 638-647
Spagnoli, A., Lucca, U., Menasce, G., Bandera, L., Cizza, G., Forloni, G., Tettamanti, M., Comelli, M.M., Long-term acetyl-L-carnitine treatment in Alzheimer's disease (1991) Neurology, 41, pp. 1726-1732
Thai, L.J., Carta, A., Clarke, W.R., Ferris, S.H., Friedland, R.P., Petersen, R.C., Pettegrew, J.W., Woolson, R.F., A 1-year multicenter placebo-controlled study of acetyl-L-carnitine in patients with Alzheimer's disease (1996) Neurology, 47 (3), pp. 705-711
Thal, L.J., Calvani, M., Amato, A., Carta, A., A 1-year controlled trial of acetyl-l-carnitine in early-onset AD (2000) Neurology, 55, pp. 805-810
Calabrese, V., Ravagna, A., Colombrita, C., Scapagnini, G., Guagliano, E., Calvani, M., Butterfield, D.A., Giuffrida Stella, A.M.G., Acetylcarnitine induces heme oxygenase in rat astrocytes and protects against oxidative stress: Involvement of the transcription factor Nrf2 (2005) Journal of Neuroscience Research, 79 (4), pp. 509-521. , DOI 10.1002/jnr.20386
Snyder, S.H., Brain peptides as neurotransmitters (1980) Science, 209 (4460), pp. 976-983
Abe, H., Role of histidine-related compounds as intracellular proton buffering constituents in vertebrate muscle (2000) Biochemistry (Mosc), 65, pp. 757-765
Hipkiss, A.R., Preston, J.E., Himsworth, D.T.M., Worthington, V.C., Keown, M., Michaelis, J., Lawrence, J., Abbott, N.J., Pluripotent protective effects of carnosine, a naturally occurring dipeptide (1998) Annals of the New York Academy of Sciences, 854, pp. 37-53. , DOI 10.1111/j.1749-6632.1998.tb09890.x
Bakardjiev, A., Biosynthesis of carnosine in primary cultures of rat olfactory bulb (1997) Neuroscience Letters, 227 (2), pp. 115-118. , DOI 10.1016/S0304-3940(97)00315-7, PII S0304394097003157
Teufel, M., Saudek, V., Ledig, J.-P., Bernhardt, A., Boularand, S., Carreau, A., Cairns, N.J., Smirnova, T., Sequence identification and characterization of human carnosinase and a closely related non-specific dipeptidase (2003) Journal of Biological Chemistry, 278 (8), pp. 6521-6531. , DOI 10.1074/jbc.M209764200
Bauer, K., X-His dipeptidase (2004) Handbook of Proteolytic Enzymes, , Eds: A. J. Barret, N. D. Rawlings, J. F. Woessner Elsevier, Amsterdam
Bauer, K., Cytosol non-specific dipeptidase (2004) Handbook of Proteolytic Enzymes, , Eds: A. J. Barret, N. D. Rawlings, J. F. Woessner, Elsevier, Amsterdam
De Marchis, S., Modena, C., Peretto, P., Giffard, C., Fasolo, A., Carnosine-like immunoreactivity in the central nervous system of rats during postnatal development (2000) J Comp Neurol, 426, pp. 378-390
Bauer, K., Carnosine and homocarnosine, the forgotten, enigmatic peptides of the brain (2005) Neurochemical Research, 30 (10), pp. 1339-1345. , DOI 10.1007/s11064-005-8806-z
Mcfarland, G.A., Holliday, R., Retardation of the senescence of cultured human diploid fibroblasts by carnosine (1994) Experimental Cell Research, 212 (2), pp. 167-175. , DOI 10.1006/excr.1994.1132
Yuneva, A.O., Kramarenko, G.G., Vetreshchak, T.V., Gallant, S., Boldyrev, A.A., Effect of carnosine on Drosophila melanogaster lifespan (2002) Bulletin of Experimental Biology and Medicine, 133 (6), pp. 559-561. , DOI 10.1023/A:1020273506970
Yuneva, M.O., Bulygina, E.R., Gallant, S.C., Kramarenko, G.G., Stvolinsky, S.L., Semyonova, M.L., Boldyrev, A.A., Effect of carnosine on age-induced changes in senescence-accelerated mice (1999) Journal of Anti-Aging Medicine, 2 (4), pp. 337-342
Babizhayev, M.A., Biological activities of the natural imidazole-containing peptidomimetics n-acetylcarnosine, carcinine and l-carnosine in ophthalmic and skin care products (2006) Life Sci, 78, pp. 2343-2357
Lee, Y.-T., Hsu, C.-C., Lin, M.-H., Liu, K.-S., Yin, M.-C., Histidine and carnosine delay diabetic deterioration in mice and protect human low density lipoprotein against oxidation and glycation (2005) European Journal of Pharmacology, 513 (1-2), pp. 145-150. , DOI 10.1016/j.ejphar.2005.02.010, PII S001429990500172X
Rashid, I., Van Reyk, D.M., Davies, M.J., Carnosine and its constituents inhibit glycation of low-density lipoproteins that promotes foam cell formation in vitro (2007) FEBS Letts, 581, pp. 1067-1070
Sauerhofer, S., Yuan, G., Braun, G.S., Deinzer, M., Neumaier, M., Gretz, N., Floege, J., Moeller, M.J., L-carnosine, a substrate of carnosinase-1, influences glucose metabolism (2007) Diabetes, 56 (10), pp. 2425-2432. , http://diabetes.diabetesjournals.org/cgi/reprint/56/10/2425, DOI 10.2337/db07-0177
Hipkiss, A.R., Glycation, ageing and carnosine: Are carnivorous diets beneficial? (2005) Mechanisms of Ageing and Development, 126 (10), pp. 1034-1039. , DOI 10.1016/j.mad.2005.05.002, PII S0047637405001193
Huang, Y., Duan, J., Chen, H., Chen, M., Chen, G., Separation and determination of carnosine-related peptides using capillary electrophoresis with laser-induced fluorescence detection (2005) Electrophoresis, 26 (3), pp. 593-599. , DOI 10.1002/elps.200406130
Hipkiss, A.R., Could carnosine suppress zinc-mediated proteasome inhibition and neurodegeneration? Therapeutic potential of a non-toxic but non-patentable dipeptide (2005) Biogerontology, 6 (2), pp. 147-149. , DOI 10.1007/s10522-005-3460-z
Dobrota, D., Fedorova, T., Stvolinsky, S., Babusikova, E., Likavcanova, K., Drgova, A., Strapkova, A., Boldyrev, A., Carnosine protects the brain of rats and mongolian gerbils against ischemic injury: After-stroke-effect (2005) Neurochemical Research, 30 (10), pp. 1283-1288. , DOI 10.1007/s11064-005-8799-7
Stvolinsky, S., Kukley, M., Dobrota, D., Mezesova, V., Boldyrev, A., Carnosine protects rats under global ischemia (2000) Brain Research Bulletin, 53 (4), pp. 445-448. , DOI 10.1016/S0361-9230(00)00366-X, PII S036192300000366X
Tang, S.-C., Arumugam, T.V., Cutler, R.G., Jo, D.-G., Magnus, T., Chan, S.L., Mughal, M.R., Mattson, M.P., Neuroprotective actions of a histidine analogue in models of ischemic stroke (2007) Journal of Neurochemistry, 101 (3), pp. 729-736. , DOI 10.1111/j.1471-4159.2006.04412.x
Pubill, D., Verdaguer, E., Sureda, F.X., Camins, A., Pallas, M., Camarasa, J., Escubedo, E., Carnosine prevents methamphetamine-induced gliosis but not dopamine terminal loss in rats (2002) European Journal of Pharmacology, 448 (2-3), pp. 165-168. , DOI 10.1016/S0014-2999(02)01949-0, PII S0014299902019490
Dukic-Stefanovic, S., Schinzel, R., Riederer, P., Munch, G., AGES in the brain ageing: AGE-inhibitors as neuroprotective and anti-dementia drugs? (2001) Biogerontology, 2 (1), pp. 19-34. , DOI 10.1023/A:1010052800347
Trombley, P.Q., Horning, M.S., Blakemore, L.J., Carnosine modulates zinc and copper effects on amino acid receptors and synaptic transmission (1998) NeuroReport, 9 (15), pp. 3503-3507
Preston, J.E., Hipkiss, A.R., Himsworth, D.T.J., Romero, I.A., Abbott, J.N., Toxic effects of β-amyloid(25-35) on immortalised rat brain endothelial cell: Protection by carnosine, homocarnosine and β-alanine (1998) Neuroscience Letters, 242 (2), pp. 105-108. , DOI 10.1016/S0304-3940(98)00058-5, PII S0304394098000585
Fu, Q., Dai, H., Hu, W., Fan, Y., Zhang, Y., Chen, Z., Carnosine protects against Aβ-42-induced neurotoxicity in differentiated rat PC12 cells Cell Mol Neurobiol, , Nov. 20
Shen, Y., Hu, W.-w., Fan, Y.-y., Dai, H.-b., Fu, Q.-l., Wei, E.-Q., Luo, J.-h., Chen, Z., Carnosine protects against NMDA-induced neurotoxicity in differentiated rat PC12 cells through carnosine-histidine-histamine pathway and H 1/H3 receptors (2007) Biochemical Pharmacology, 73 (5), pp. 709-717. , DOI 10.1016/j.bcp.2006.11.007, PII S0006295206007428
Fonteh, A.N., Harrington, R.J., Tsai, A., Liao, P., Harrington, M.G., Free amino acid and dipeptide changes in the body fluids from Alzheimer's disease subjects (2007) Amino Acids, 32 (2), pp. 213-224. , DOI 10.1007/s00726-006-0409-8, Special Issue: Focus on Metabotropic Glutamate Receptors
Fontana, M., Pinnen, F., Lucente, G., Pecci, L., Prevention of peroxynitrite-dependent damage by carnosine and related sulphonamido pseudodipeptides (2002) Cellular and Molecular Life Sciences, 59 (3), pp. 546-551. , DOI 10.1007/s00018-002-8446-2
Severina, I.S., Bussygina, O.G., Pyatakova, N.V., Carnosine as a regulator of soluble guanylate cyclase (2000) Biochemistry (Mosc), 65, pp. 783-788
Nicoletti, V.G., Santoro, A.M., Grasso, G., Vagliasindi, L.I., Giuffrida, M.L., Cuppari, C., Purrello, V.S., Rizzarelli, E., Carnosine interaction with nitric oxide and astroglial cell protection (2007) Journal of Neuroscience Research, 85 (10), pp. 2239-2245. , DOI 10.1002/jnr.21365
La Mendola, D., Sortino, S., Vecchio, G., Rizzarelli, E., Synthesis of new carnosine derivatives of β-cyclodextrin and their hydroxyl radical scavenger ability (2002) Helvetica Chimica Acta, 85 (6), pp. 1633-1643. , DOI 10.1002/1522-2675(200206)85:6<1633::AID-HLCA1633>
3.0.CO
Bonomo, R.P., Bruno, V., Conte, E., De Guidi, G., La Mendola, D., Maccarrone, G., Nicoletti, F., Vecchio, G., Potentiometric, spectroscopic and antioxidant activity studies of SOD mimics containing carnosine (2003) J Chem Soc Dalton Trans, pp. 4406-4415
Mineo, P., Vitalini, D., La Mendola, D., Rizzarelli, E., Scamporrino, E., Vecchio, G., Coordination features of difunctionalized β-cyclodextrins with carnosine: ESI-MS and spectroscopic investigations on 6A,6D-di-(β-alanyl-L- histidine)-6A,6D-dideoxy-β-cyclodextrin and 6A,6C-di-(β-alanyl-L- histidine)-6A,6C-dideoxy-β-cyclodextrin and their copper(II) complexes (2004) Journal of Inorganic Biochemistry, 98 (2), pp. 254-265. , DOI 10.1016/j.jinorgbio.2003.10.015
Bellia, F., La Mendola, D., Maccarrone, G., Mineo, P., Vitalizi, D., Scamporrino, E., Sortino, S., Rizzarelli, E., Copper (II) complexes with β-cyclodextrinhomocarnosine conjugates and their antioxidant activity (2007) Inorg Chim Acta, 360, pp. 949-954
Amorini, A.M., Bellia, F., Di Pietro, V., Giardina, B., La Mendola, D., Lazzarino, G., Sortino, S., Vecchio, G., Synthesis and antioxidant activity of new homocarnosine β-cyclodextrin conjugates (2007) European Journal of Medicinal Chemistry, 42 (7), pp. 910-920. , DOI 10.1016/j.ejmech.2006.12.036, PII S0223523407000384
Bellia, F., Amorini, A.M., La Mendola, D., Vecchio, G., Gavazzi, B., Giardina, B., Di Pietro, V., Rizzarelli, E., New glycosidic derivatives of histidinecontaining dipeptides with antioxidant properties and resistant to carnosinase activity (2008) Eur J Med Chem, , in press
Adlard, P.A., Bush, A.I., Metals and Alzheimer's disease (2006) Journal of Alzheimer's Disease, 10 (2-3), pp. 145-163
Chrouch, P.J., White, A.R., Bush, A.I., The modulation of metal bio-availability as a therapeutic strategy for the treatment of Alzheimer's disease (2007) FEBS J, 274, pp. 375-3783
Mattson, M.P., Hormesis defined (2008) Ageing Res Rev, 7, pp. 1-7
Calabrese, E.J., Baldwin, L.A., Hormesis as a biological hypothesis (1998) Environmental Health Perspectives, 106 (SUPPL. 1), pp. 357-362
Rattan, S.I., The nature of gerontogenes and vitagenes. Antiaging effects of repeated heat shock on human fibroblasts (1998) Ann N Y Acad Sci, 854, pp. 54-60
Calabrese, V., Stella, A.M.G., Calvani, M., Butterfield, D.A., Acetylcarnitine and cellular stress response: Roles in nutritional redox homeostasis and regulation of longevity genes (2006) Journal of Nutritional Biochemistry, 17 (2), pp. 73-88. , DOI 10.1016/j.jnutbio.2005.03.027, PII S0955286305002433
Mancuso, C., Bates, T.E., Butterfield, D.A., Calafato, S., Cornelius, C., De Lorenzo, A., Dinkova Kostova, A.T., Calabrese, V., Natural antioxidants in Alzheimer's disease (2007) Expert Opinion on Investigational Drugs, 16 (12), pp. 1921-1931. , DOI 10.1517/13543784.16.12.1921
Vitagenes, dietary antioxidants and neuroprotection in neurodegenerative diseases
The ability of a cell to counteract stressful conditions, known as cellular stress response, requires the activation of pro-survival pathways and the production of molecules with anti-oxidant, anti-apoptotic or pro-apoptotic activities. Among the cellular pathways conferring protection against oxidative stress, a key role is played by vitagenes, which include heat shock proteins (Hsps) heme oxygenase-1 and Hsp70, as well as the thioredoxin/thioredoxin reductase system. Heat shock response contributes to establish a cytoprotective state in a wide variety of human diseases, including inflammation, cancer, aging and neurodegenerative disorders. Given the broad cytoprotective properties of the heat shock response there is now strong interest in discovering and developing pharmacological agents capable of inducing stress responses. Dietary antioxidants, such as curcumin, L-carnitine/acetyl-L-carnitine and carnosine have recently been demonstrated in vitro to be neuroprotective through the activation of hormetic pathways, including vitagenes. In the present review we discuss the importance of vitagenes in the cellular stress response and analyse, from a pharmacological point of view, the potential use of dietary antioxidants in the treatment of neurodegenerative disorders in humans.
Vitagenes, dietary antioxidants and neuroprotection in neurodegenerative diseases
No results.
Vitagenes, dietary antioxidants and neuroprotection in neurodegenerative diseases
Ciccarelli M, Sorriento D, Coscioni E, Iaccarino G, Santulli G * Adrenergic Receptors(262 views) Endocrinol Of The Heart In Health And Dis (ISSN: 9780-1280311249780128031117), 2016; N/D: 285-315. Impact Factor:0 ViewExport to BibTeXExport to EndNote
Kim YH, Shin SW, Pellicano R, Fagoonee S, Choi IJ, Kim YI, Park B, Choi JM, Kim SG, Choi J, Park JY, Oh S, Yang HJ, Lim JH, Im JP, Kim JS, Jung HC, Ponzetto A, Figura N, Malfertheiner P, Choi IJ, Kook MC, Kim YI, Cho SJ, Lee JY, Kim CG, Park B, Nam BH, Bae SE, Choi KD, Choe J, Kim SO, Na HK, Choi JY, Ahn JY, Jung KW, Lee J, Kim DH, Chang HS, Song HJ, Lee GH, Jung HY, Seta T, Takahashi Y, Noguchi Y, Shikata S, Sakai T, Sakai K, Yamashita Y, Nakayama T, Leja M, Park JY, Murillo R, Liepniece-karele I, Isajevs S, Kikuste I, Rudzite D, Krike P, Parshutin S, Polaka I, Kirsners A, Santare D, Folkmanis V, Daugule I, Plummer M, Herrero R, Tsukamoto T, Nakagawa M, Kiriyama Y, Toyoda T, Cao X, Corral JE, Mera R, Dye CW, Morgan DR, Lee YC, Lin JT, Garcia Martin R, Matia Cubillo A, Lee SH, Park JM, Han YM, Ko WJ, Hahm KB, Leontiadis GI, Ford AC, Ichinose M, Sugano K, Jeong M, Park JM, Han YM, Park KY, Lee DH, Yoo JH, Cho JY, Hahm KB, Bang CS, Baik GH, Shin IS, Kim JB, Suk KT, Yoon JH, Kim YS, Kim DJ * Helicobacter pylori Eradication for Prevention of Metachronous Recurrence after Endoscopic Resection of Early Gastric Cancer(297 views) N Engl J Med (ISSN: 0028-4793, 0028-4793linking, 1533-4406electronic), 2015 Jun; 30642104201566393291: 749-756. Impact Factor:59.558 ViewExport to BibTeXExport to EndNote