메뉴 건너뛰기




Volumn 2014, Issue , 2014, Pages

Lipid peroxidation: Production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal

Author keywords

[No Author keywords available]

Indexed keywords

ALDEHYDES; CELL DEATH; FREE RADICALS; GENE EXPRESSION; LIPIDS; MAMMALS; METABOLISM; OXIDATIVE STRESS; PHYSIOLOGY; POLYUNSATURATED FATTY ACIDS;

EID: 84901917562     PISSN: 19420900     EISSN: 19420994     Source Type: Journal    
DOI: 10.1155/2014/360438     Document Type: Review
Times cited : (4252)

References (434)
  • 4
    • 80053370162 scopus 로고    scopus 로고
    • Lipidomics of polyunsaturated-fatty-acid-derived oxygenated metabolites
    • 2-s2.0-80053370162 10.1042/BST0391240
    • Massey K. A., Nicolaou A., Lipidomics of polyunsaturated-fatty-acid- derived oxygenated metabolites. Biochemical Society Transactions 2011 39 5 1240 1246 2-s2.0-80053370162 10.1042/BST0391240
    • (2011) Biochemical Society Transactions , vol.39 , Issue.5 , pp. 1240-1246
    • Massey, K.A.1    Nicolaou, A.2
  • 6
    • 84860497397 scopus 로고    scopus 로고
    • Diacylglycerol activation of protein kinase C ε and hepatic insulin resistance
    • 2-s2.0-84860497397 10.1016/j.cmet.2012.03.005
    • Jornayvaz F. R., Shulman G. I., Diacylglycerol activation of protein kinase C ε and hepatic insulin resistance. Cell Metabolism 2012 15 5 574 584 2-s2.0-84860497397 10.1016/j.cmet.2012.03.005
    • (2012) Cell Metabolism , vol.15 , Issue.5 , pp. 574-584
    • Jornayvaz, F.R.1    Shulman, G.I.2
  • 8
    • 34147138401 scopus 로고    scopus 로고
    • Chimaerins: GAPs that bridge diacylglycerol signalling and the small G-protein Rac
    • DOI 10.1042/BJ20061750
    • Yang C., Kazanietz M. G., Chimaerins: GAPs that bridge diacylglycerol signalling and the small G-protein Rac. Biochemical Journal 2007 403 1 1 12 (Pubitemid 46569861)
    • (2007) Biochemical Journal , vol.403 , Issue.1 , pp. 1-12
    • Yang, C.1    Kazanietz, M.G.2
  • 10
    • 0036677944 scopus 로고    scopus 로고
    • Inositol and higher inositol phosphates in neural tissues: Homeostasis, metabolism and functional significance
    • 2-s2.0-0036677944 10.1046/j.1471-4159.2002.01041.x
    • Fisher S. K., Novak J. E., Agranoff B. W., Inositol and higher inositol phosphates in neural tissues: homeostasis, metabolism and functional significance. Journal of Neurochemistry 2002 82 4 736 754 2-s2.0-0036677944 10.1046/j.1471-4159.2002.01041.x
    • (2002) Journal of Neurochemistry , vol.82 , Issue.4 , pp. 736-754
    • Fisher, S.K.1    Novak, J.E.2    Agranoff, B.W.3
  • 11
    • 34547102761 scopus 로고    scopus 로고
    • Biology-enabling inositol phosphates, phosphatidylinositol phosphates and derivatives
    • DOI 10.1039/b407701f
    • Conway S. J., Miller G. J., Biology-enabling inositol phosphates, phosphatidylinositol phosphates and derivatives. Natural Product Reports 2007 24 4 687 707 2-s2.0-34547102761 10.1039/b407701f (Pubitemid 47106911)
    • (2007) Natural Product Reports , vol.24 , Issue.4 , pp. 687-707
    • Conway, S.J.1    Miller, G.J.2
  • 12
    • 84867713914 scopus 로고    scopus 로고
    • Sphingosine-1-phosphate signaling in physiology and diseases
    • Takuwa Y., Okamoto Y., Yoshioka K., Takuwa N., Sphingosine-1-phosphate signaling in physiology and diseases. BioFactors 2012 38 5 329 337
    • (2012) BioFactors , vol.38 , Issue.5 , pp. 329-337
    • Takuwa, Y.1    Okamoto, Y.2    Yoshioka, K.3    Takuwa, N.4
  • 14
    • 38549152194 scopus 로고    scopus 로고
    • Principles of bioactive lipid signalling: Lessons from sphingolipids
    • DOI 10.1038/nrm2329, PII NRM2329
    • Hannun Y. A., Obeid L. M., Principles of bioactive lipid signalling: lessons from sphingolipids. Nature Reviews Molecular Cell Biology 2008 9 2 139 150 (Pubitemid 351158911)
    • (2008) Nature Reviews Molecular Cell Biology , vol.9 , Issue.2 , pp. 139-150
    • Hannun, Y.A.1    Obeid, L.M.2
  • 16
    • 84857053651 scopus 로고    scopus 로고
    • Anti-inflammation therapy by activation of prostaglandin EP4 receptor in cardiovascular and other inflammatory diseases
    • 2-s2.0-84857053651 10.1097/FJC.0b013e3182244a12
    • Tang E. H. C., Libby P., Vanhoutte P. M., Xu A., Anti-inflammation therapy by activation of prostaglandin EP4 receptor in cardiovascular and other inflammatory diseases. Journal of Cardiovascular Pharmacology 2012 59 2 116 123 2-s2.0-84857053651 10.1097/FJC.0b013e3182244a12
    • (2012) Journal of Cardiovascular Pharmacology , vol.59 , Issue.2 , pp. 116-123
    • Tang, E.H.C.1    Libby, P.2    Vanhoutte, P.M.3    Xu, A.4
  • 20
    • 8644255769 scopus 로고    scopus 로고
    • Oxygen free radicals and redox biology of organelles
    • DOI 10.1007/s00418-004-0676-y
    • Moldovan L., Moldovan N. I., Oxygen free radicals and redox biology of organelles. Histochemistry and Cell Biology 2004 122 4 395 412 2-s2.0-8644255769 10.1007/s00418-004-0676-y (Pubitemid 39506596)
    • (2004) Histochemistry and Cell Biology , vol.122 , Issue.4 , pp. 395-412
    • Moldovan, L.1    Moldovan, N.I.2
  • 22
    • 0021351203 scopus 로고
    • Oxygen toxicity, oxygen radicals, transition metals and disease
    • Halliwell B., Gutteridge J. M. C., Oxygen toxicity, oxygen radicals, transition metals and disease. Biochemical Journal 1984 219 1 1 14 2-s2.0-0021351203 (Pubitemid 14171443)
    • (1984) Biochemical Journal , vol.219 , Issue.1 , pp. 1-14
    • Halliwell, B.1    Gutteridge, J.M.C.2
  • 23
    • 0037220471 scopus 로고    scopus 로고
    • Evidence for dopamine-derived hydroxyl radical formation in the nigrostriatal system in response to axotomy
    • DOI 10.1016/S0891-5849(02)01191-7, PII S0891584902011917
    • Venero J. L., Revuelta M., Atiki L., Santiago M., Tomás-Camardiel M. C., Cano J., Machado A., Evidence for dopamine-derived hydroxyl radical formation in the nigrostriatal system in response to axotomy. Free Radical Biology and Medicine 2003 34 1 111 123 2-s2.0-0037220471 10.1016/S0891-5849(02) 01191-7 (Pubitemid 36043892)
    • (2003) Free Radical Biology and Medicine , vol.34 , Issue.1 , pp. 111-123
    • Venero, J.L.1    Revuelta, M.2    Atiki, L.3    Santiago, M.4    Tomas-Camardiel, M.C.5    Cano, J.6    Machado, A.7
  • 24
    • 3042838351 scopus 로고    scopus 로고
    • Contribution of redox-active iron and copper to oxidative damage in Alzheimer disease
    • DOI 10.1016/j.arr.2004.01.002, PII S1568163704000133
    • Castellani R. J., Honda K., Zhu X., Cash A. D., Nunomura A., Perry G., Smith M. A., Contribution of redox-active iron and copper to oxidative damage in Alzheimer disease. Ageing Research Reviews 2004 3 3 319 326 2-s2.0-3042838351 10.1016/j.arr.2004.01.002 (Pubitemid 38849409)
    • (2004) Ageing Research Reviews , vol.3 , Issue.3 , pp. 319-326
    • Castellani, R.J.1    Honda, K.2    Zhu, X.3    Cash, A.D.4    Nunomura, A.5    Perry, G.6    Smith, M.A.7
  • 25
    • 84866080059 scopus 로고    scopus 로고
    • Hydroxyl radical-modified fibrinogen as a marker of thrombosis: The role of iron
    • Lipinski B., Pretorius E., Hydroxyl radical-modified fibrinogen as a marker of thrombosis: the role of iron. Hematology 2012 17 4 241 247
    • (2012) Hematology , vol.17 , Issue.4 , pp. 241-247
    • Lipinski, B.1    Pretorius, E.2
  • 26
    • 84858133452 scopus 로고    scopus 로고
    • Mechanisms of free radical-induced damage to DNA
    • 2-s2.0-84858133452 10.3109/10715762.2011.653969
    • Dizdaroglu M., Jaruga P., Mechanisms of free radical-induced damage to DNA. Free Radical Research 2012 46 4 382 419 2-s2.0-84858133452 10.3109/10715762.2011.653969
    • (2012) Free Radical Research , vol.46 , Issue.4 , pp. 382-419
    • Dizdaroglu, M.1    Jaruga, P.2
  • 27
    • 84863442754 scopus 로고    scopus 로고
    • Literature review of the role of hydroxyl radicals in chemically-induced mutagenicity and carcinogenicity for the risk assessment of a disinfection system utilizing photolysis of hydrogen peroxide
    • Kanno T., Nakamura K., Ikai H., Kikuchi K., Sasaki K., Niwano Y., Literature review of the role of hydroxyl radicals in chemically-induced mutagenicity and carcinogenicity for the risk assessment of a disinfection system utilizing photolysis of hydrogen peroxide. Journal of Clinical Biochemistry and Nutrition 2012 51 1 9 14
    • (2012) Journal of Clinical Biochemistry and Nutrition , vol.51 , Issue.1 , pp. 9-14
    • Kanno, T.1    Nakamura, K.2    Ikai, H.3    Kikuchi, K.4    Sasaki, K.5    Niwano, Y.6
  • 28
    • 0020646527 scopus 로고
    • A study of the reactivity of HO2/O2- with unsaturated fatty acids
    • 2-s2.0-0020646527
    • Bielski B. H. J., Arudi R. L., Sutherland M. W., A study of the reactivity of HO2/O2- with unsaturated fatty acids. Journal of Biological Chemistry 1983 258 8 4759 4761 2-s2.0-0020646527
    • (1983) Journal of Biological Chemistry , vol.258 , Issue.8 , pp. 4759-4761
    • Bielski, B.H.J.1    Arudi, R.L.2    Sutherland, M.W.3
  • 29
    • 43149115625 scopus 로고    scopus 로고
    • Intermolecular peroxyl radical reactions during autoxidation of hydroxy and hydroperoxy arachidonic acids generate a novel series of epoxidized products
    • DOI 10.1021/tx700357u
    • Schneider C., Boeglin W. E., Yin H., Porter N. A., Brash A. R., Intermolecular peroxyl radical reactions during autoxidation of hydroxy and hydroperoxy arachidonic acids generate a novel series of epoxidized products. Chemical Research in Toxicology 2008 21 4 895 903 2-s2.0-43149115625 10.1021/tx700357u (Pubitemid 351644568)
    • (2008) Chemical Research in Toxicology , vol.21 , Issue.4 , pp. 895-903
    • Schneider, C.1    Boeglin, W.E.2    Yin, H.3    Porter, N.A.4    Brash, A.R.5
  • 30
    • 0034044702 scopus 로고    scopus 로고
    • HPLC analysis of lipid-derived polyunsaturated fatty acid peroxidation products in oxidatively modified human plasma
    • Browne R. W., Armstrong D., HPLC analysis of lipid-derived polyunsaturated fatty acid peroxidation products in oxidatively modified human plasma. Clinical Chemistry 2000 46 6, part 1 829 836 2-s2.0-0034044702 (Pubitemid 30397447)
    • (2000) Clinical Chemistry , vol.46 , Issue.6 , pp. 829-836
    • Browne, R.W.1    Armstrong, D.2
  • 31
    • 80054089650 scopus 로고    scopus 로고
    • Free radical lipid peroxidation: Mechanisms and analysis
    • 2-s2.0-80054089650 10.1021/cr200084z
    • Yin H., Xu L., Porter N. A., Free radical lipid peroxidation: mechanisms and analysis. Chemical Reviews 2011 111 10 5944 5972 2-s2.0-80054089650 10.1021/cr200084z
    • (2011) Chemical Reviews , vol.111 , Issue.10 , pp. 5944-5972
    • Yin, H.1    Xu, L.2    Porter, N.A.3
  • 32
    • 84878761873 scopus 로고    scopus 로고
    • Oxidized phosphatidylcholines in membrane-level cellular signaling: From biophysics to physiology and molecular pathology
    • Volinsky R., Kinnunen P. K. J., Oxidized phosphatidylcholines in membrane-level cellular signaling: from biophysics to physiology and molecular pathology. FEBS Journal 2013 280 12 2806 2816
    • (2013) FEBS Journal , vol.280 , Issue.12 , pp. 2806-2816
    • Volinsky, R.1    Kinnunen, P.K.J.2
  • 33
    • 84863990492 scopus 로고    scopus 로고
    • Protein-oxidized phospholipid interactions in cellular signaling for cell death: From biophysics to clinical correlations
    • Kinnunen P. K. J., Kaarniranta K., Mahalka A. K., Protein-oxidized phospholipid interactions in cellular signaling for cell death: from biophysics to clinical correlations. Biochimica et Biophysica Acta 2012 1818 10 2446 2455
    • (2012) Biochimica et Biophysica Acta , vol.1818 , Issue.10 , pp. 2446-2455
    • Kinnunen, P.K.J.1    Kaarniranta, K.2    Mahalka, A.K.3
  • 34
    • 84863989944 scopus 로고    scopus 로고
    • Chemistry of phospholipid oxidation
    • Reis A., Spickett C. M., Chemistry of phospholipid oxidation. Biochimica et Biophysica Acta 2012 1818 10 2374 2387
    • (2012) Biochimica et Biophysica Acta , vol.1818 , Issue.10 , pp. 2374-2387
    • Reis, A.1    Spickett, C.M.2
  • 35
    • 34249991689 scopus 로고    scopus 로고
    • Oxidized phospholipids: From molecular properties to disease
    • DOI 10.1016/j.bbadis.2007.04.009, PII S0925443907001032
    • Fruhwirth G. O., Loidl A., Hermetter A., Oxidized phospholipids: from molecular properties to disease. Biochimica et Biophysica Acta: Molecular Basis of Disease 2007 1772 7 718 736 2-s2.0-34249991689 10.1016/j.bbadis.2007.04.009 (Pubitemid 46891216)
    • (2007) Biochimica et Biophysica Acta - Molecular Basis of Disease , vol.1772 , Issue.7 , pp. 718-736
    • Fruhwirth, G.O.1    Loidl, A.2    Hermetter, A.3
  • 36
    • 0031821531 scopus 로고    scopus 로고
    • Lipid hydroperoxide generation, turnover, and effector action in biological systems
    • Girotti A. W., Lipid hydroperoxide generation, turnover, and effector action in biological systems. Journal of Lipid Research 1998 39 8 1529 1542 2-s2.0-0031821531 (Pubitemid 28377917)
    • (1998) Journal of Lipid Research , vol.39 , Issue.8 , pp. 1529-1542
    • Girotti, A.W.1
  • 38
    • 0020365443 scopus 로고
    • 2+ in rat liver microsomes
    • Esterbauer H., Cheeseman K. H., Dianzani M. U., Separation and characterization of the aldehydic products of lipid peroxidation stimulated by ADP-Fe2+ in rat liver microsomes. Biochemical Journal 1982 208 1 129 140 2-s2.0-0020365443 (Pubitemid 13215400)
    • (1982) Biochemical Journal , vol.208 , Issue.1 , pp. 129-140
    • Esterbauer, H.1    Cheeseman, K.H.2    Dianzani, M.U.3
  • 39
    • 0021842533 scopus 로고
    • Separation and characterization of the aldehydic products of lipid peroxidation stimulated by carbon tetrachloride or ADP-iron in isolated rat hepatocytes and rat liver microsomal suspensions
    • Poli G., Dianzani M. U., Cheeseman K. H., Slater T. F., Lang J., Esterbauer H., Separation and characterization of the aldehydic products of lipid peroxidation stimulated by carbon tetrachloride or ADP-iron in isolated rat hepatocytes and rat liver microsomal suspensions. Biochemical Journal 1985 227 2 629 638 2-s2.0-0021842533 (Pubitemid 15067738)
    • (1985) Biochemical Journal , vol.227 , Issue.2 , pp. 629-638
    • Poli, G.1    Dianzani, M.U.2    Cheeseman, K.H.3
  • 40
    • 0019193338 scopus 로고
    • Identification of 4-hydroxynonenal as a cytotoxic product originating from the peroxidation of liver microsomal lipids
    • Benedetti A., Comporti M., Esterbauer H., Identification of 4-hydroxynonenal as a cytotoxic product originating from the peroxidation of liver microsomal lipids. Biochimica et Biophysica Acta 1980 620 2 281 296 2-s2.0-0019193338 (Pubitemid 11216247)
    • (1980) Biochimica et Biophysica Acta , vol.620 , Issue.2 , pp. 281-296
    • Benedetti, A.1    Comporti, M.2    Esterbauer, H.3
  • 41
    • 0020510128 scopus 로고
    • Effects of 4-hydroxynonenal on isolated hepatocytes. Studies on chemiluminescence response, alkane production and glutathione status
    • Cadenas E., Müller A., Brigelius R., Esterbauer H., Sies H., Effects of 4-hydroxynonenal on isolated hepatocytes. Studies on chemiluminescence response, alkane production and glutathione status. Biochemical Journal 1983 214 2 479 487 2-s2.0-0020510128 (Pubitemid 13035530)
    • (1983) Biochemical Journal , vol.214 , Issue.2 , pp. 479-487
    • Cadenas, E.1    Muller, A.2    Brigelius, R.3
  • 42
    • 0021288842 scopus 로고
    • Detection of malonaldehyde by high-performance liquid chromatography
    • Esterbauer H., Lang J., Zadravec S., Slater T. F., Detection of malonaldehyde by high-performance liquid chromatography. Methods in Enzymology 1984 105 319 328 2-s2.0-0021288842 (Pubitemid 14165393)
    • (1984) Methods in Enzymology , vol.VOL. 105 , pp. 319-328
    • Esterbauer, H.1    Lang, J.2    Zadravec, S.3    Slater, T.F.4
  • 43
    • 0021704943 scopus 로고
    • Detection of 4-hydroxynonenal as a product of lipid peroxidation in native Ehrlich ascites tumor cells
    • DOI 10.1016/0005-2760(84)90122-X
    • Winkler P., Lindner W., Esterbauer H., Schauenstein E., Schaur R. J., Khoschsorur G. A., Detection of 4-hydroxynonenal as a product of lipid peroxidation in native Ehrlich ascites tumor cells. Biochimica et Biophysica Acta: Lipids and Lipid Metabolism 1984 796 3 232 237 2-s2.0-0021704943 10.1016/0005-2760(84)90122-X (Pubitemid 15158769)
    • (1984) Biochimica et Biophysica Acta - Lipids and Lipid Metabolism , vol.796 , Issue.3 , pp. 232-237
    • Winkler, P.1    Lindner, W.2    Esterbauer, H.3
  • 44
    • 0022548096 scopus 로고
    • Studies on the mechanism of formation of 4-hydroxynonenal during microsomal lipid peroxidation
    • DOI 10.1016/0005-2760(86)90329-2
    • Esterbauer H., Benedetti A., Lang J., Fulceri R., Fauler G., Comporti M., Studies on the mechanism of formation of 4-hydroxynonenal during microsomal lipid peroxidation. Biochimica et Biophysica Acta: Lipids and Lipid Metabolism 1986 876 1 154 166 2-s2.0-0022548096 (Pubitemid 16011009)
    • (1986) Biochimica et Biophysica Acta - Lipids and Lipid Metabolism , vol.876 , Issue.1 , pp. 154-166
    • Esterbauer, H.1    Benedetti, A.2    Lang, J.3
  • 45
    • 0023150756 scopus 로고
    • Effects of the lipid peroxidation product 4-hydroxynonenal on the aggregation of human platelets
    • DOI 10.1016/0009-2797(87)90033-0
    • Hurst J. S., Slater T. F., Lang J., Effects of the lipid peroxidation product 4-hydroxynonenal on the aggregation of human platelets. Chemico-Biological Interactions 1987 61 2 109 124 2-s2.0-0023150756 (Pubitemid 17056445)
    • (1987) Chemico-Biological Interactions , vol.61 , Issue.2 , pp. 109-124
    • Hurst, J.S.1    Slater, T.F.2    Lang, J.3
  • 46
    • 0023916221 scopus 로고
    • Hydroxyl-radical-induced iron-catalysed degradation of 2-deoxyribose. Quantitative determination of malondialdehyde
    • 2-s2.0-0023916221
    • Cheeseman K. H., Beavis A., Esterbauer H., Hydroxyl-radical-induced iron-catalysed degradation of 2-deoxyribose. Quantitative determination of malondialdehyde. Biochemical Journal 1988 252 3 649 653 2-s2.0-0023916221
    • (1988) Biochemical Journal , vol.252 , Issue.3 , pp. 649-653
    • Cheeseman, K.H.1    Beavis, A.2    Esterbauer, H.3
  • 47
    • 0024468045 scopus 로고
    • Methods for determination of aldehydic lipid peroxidation products
    • DOI 10.1016/0891-5849(89)90015-4
    • Esterbauer H., Zolliner H., Methods for determination of aldehydic lipid peroxidation products. Free Radical Biology and Medicine 1989 7 2 197 203 2-s2.0-0024468045 (Pubitemid 19220721)
    • (1989) Free Radical Biology and Medicine , vol.7 , Issue.2 , pp. 197-203
    • Esterbauer, H.1    Zolliner, H.2
  • 48
    • 0025127633 scopus 로고
    • Determination of aldehydic lipid peroxidation products: Malonaldehyde and 4-hydroxynonenal
    • DOI 10.1016/0076-6879(90)86134-H
    • Esterbauer H., Cheeseman K. H., Determination of aldehydic lipid peroxidation products: malonaldehyde and 4-hydroxynonenal. Methods in Enzymology 1990 186 407 421 2-s2.0-0025127633 10.1016/0076-6879(90)86134-H (Pubitemid 20279982)
    • (1990) Methods in Enzymology , vol.186 , pp. 407-421
    • Esterbauer, H.1    Cheeseman, K.H.2
  • 49
    • 0025814980 scopus 로고
    • Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes
    • Esterbauer H., Schaur R. J., Zollner H., Chemistry and Biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radical Biology and Medicine 1991 11 1 81 128 2-s2.0-0025814980 10.1016/0891-5849(91)90192-6 (Pubitemid 121003917)
    • (1991) Free Radical Biology and Medicine , vol.11 , Issue.1 , pp. 81-128
    • Esterbauer, H.1    Schaur, R.J.2    Zollner, H.3
  • 50
    • 0025305055 scopus 로고
    • Possible mutagens derived from lipids and lipid precursors
    • 2-s2.0-0025305055
    • Esterbauer H., Eckl P., Ortner A., Possible mutagens derived from lipids and lipid precursors. Mutation Research 1990 238 3 223 233 2-s2.0-0025305055
    • (1990) Mutation Research , vol.238 , Issue.3 , pp. 223-233
    • Esterbauer, H.1    Eckl, P.2    Ortner, A.3
  • 51
    • 0024458152 scopus 로고
    • On the detection of lipid hydroperoxides in biological samples
    • DOI 10.1016/0891-5849(89)90010-5
    • Pryor W. A., On the detection of lipid hydroperoxides in biological samples. Free Radical Biology and Medicine 1989 7 2 177 178 2-s2.0-0024458152 (Pubitemid 19220716)
    • (1989) Free Radical Biology and Medicine , vol.7 , Issue.2 , pp. 177-178
    • Pryor, W.A.1
  • 52
    • 84982338423 scopus 로고
    • Characterization of the red pigment formed in the 2-thiobarbituric acid determination of oxidative rancidity
    • Sinnhuber R. O., Yu T. C., Yu T. C., Characterization of the red pigment formed in the 2-thiobarbituric acid determination of oxidative rancidity. Journal of Food Science 1958 23 6 626 634
    • (1958) Journal of Food Science , vol.23 , Issue.6 , pp. 626-634
    • Sinnhuber, R.O.1    Yu, T.C.2    Yu, T.C.3
  • 53
    • 84862860945 scopus 로고    scopus 로고
    • Recent advancements in the LC- and GC-based analysis of malondialdehyde (MDA): A brief overview
    • 2-s2.0-84859364419 10.1007/s10337-012-2237-1
    • Giera M., Lingeman H., Niessen W. M. A., Recent advancements in the LC- and GC-based analysis of malondialdehyde (MDA): a brief overview. Chromatographia 2012 75 9-10 433 440 2-s2.0-84859364419 10.1007/s10337-012-2237- 1
    • (2012) Chromatographia , vol.75 , Issue.9-10 , pp. 433-440
    • Giera, M.1    Lingeman, H.2    Niessen, W.M.A.3
  • 54
    • 0014127225 scopus 로고
    • Autoxidation of polyunsaturated esters in water: Chemical structure and biological activity of the products
    • 2-s2.0-0014127225
    • Schauenstein E., Autoxidation of polyunsaturated esters in water: chemical structure and biological activity of the products. Journal of Lipid Research 1967 8 5 417 428 2-s2.0-0014127225
    • (1967) Journal of Lipid Research , vol.8 , Issue.5 , pp. 417-428
    • Schauenstein, E.1
  • 55
    • 0022537754 scopus 로고
    • Cytotoxicity, DNA fragmentation and sister-chromatid exchange in Chinese hamster ovary cells exposed to the lipid peroxidation product 4-hydroxynonenal and homologous aldehydes
    • Brambilla G., Sciabà L., Faggin P., Maura A., Marinari U. M., Ferro M., Esterbauer H., Cytotoxicity, DNA fragmentation and sister-chromatid exchange in Chinese hamster ovary cells exposed to the lipid peroxidation product 4-hydroxynonenal and homologous aldehydes. Mutation Research 1986 171 2-3 169 176 2-s2.0-0022537754 (Pubitemid 16051639)
    • (1986) Mutation Research , vol.171 , Issue.2-3 , pp. 169-176
    • Brambilla, G.1    Sciaba, L.2    Faggin, P.3
  • 56
    • 0043172509 scopus 로고    scopus 로고
    • Basic aspects of the biochemical reactivity of 4-hydroxynonenal
    • DOI 10.1016/S0098-2997(03)00009-8
    • Schaur R. J., Basic aspects of the biochemical reactivity of 4-hydroxynonenal. Molecular Aspects of Medicine 2003 24 4-5 149 159 2-s2.0-0043172509 10.1016/S0098-2997(03)00009-8 (Pubitemid 36945015)
    • (2003) Molecular Aspects of Medicine , vol.24 , Issue.4-5 , pp. 149-159
    • Schaur, R.J.1
  • 57
    • 0043172469 scopus 로고    scopus 로고
    • 4-Hydroxynonenal as a bioactive marker of pathophysiological processes
    • DOI 10.1016/S0098-2997(03)00023-2
    • Zarkovic N., 4-Hydroxynonenal as a bioactive marker of pathophysiological processes. Molecular Aspects of Medicine 2003 24 4-5 281 291 2-s2.0-0043172469 10.1016/S0098-2997(03)00023-2 (Pubitemid 36945028)
    • (2003) Molecular Aspects of Medicine , vol.24 , Issue.4-5 , pp. 281-291
    • Zarkovic, N.1
  • 58
    • 84890125992 scopus 로고    scopus 로고
    • Biomarkers of lipid peroxidation in clinical material
    • Niki E., Biomarkers of lipid peroxidation in clinical material. Biochimica et Biophysica Acta 2014 1840 2 809 817
    • (2014) Biochimica et Biophysica Acta , vol.1840 , Issue.2 , pp. 809-817
    • Niki, E.1
  • 59
    • 7944227479 scopus 로고    scopus 로고
    • Do the serum oxidative stress biomarkers provide a reasonable index of the general oxidative stress status?
    • DOI 10.1016/j.bbagen.2004.06.023, PII S0304416504001710
    • Argüelles S., García S., Maldonado M., Machado A., Ayala A., Do the serum oxidative stress biomarkers provide a reasonable index of the general oxidative stress status? Biochimica et Biophysica Acta: General Subjects 2004 1674 3 251 259 2-s2.0-7944227479 10.1016/j.bbagen.2004.06.023 (Pubitemid 39469284)
    • (2004) Biochimica et Biophysica Acta - General Subjects , vol.1674 , Issue.3 , pp. 251-259
    • Arguelles, S.1    Garcia, S.2    Maldonado, M.3    Machado, A.4    Ayala, A.5
  • 60
    • 37549041636 scopus 로고    scopus 로고
    • A preliminary analysis of within-subject variation in human serum oxidative stress parameters as a function of time
    • 2-s2.0-37549041636 10.1089/rej.2006.0528
    • Argüelles S., Gómez A., Machado A., Ayala A., A preliminary analysis of within-subject variation in human serum oxidative stress parameters as a function of time. Rejuvenation Research 2007 10 4 621 636 2-s2.0-37549041636 10.1089/rej.2006.0528
    • (2007) Rejuvenation Research , vol.10 , Issue.4 , pp. 621-636
    • Argüelles, S.1    Gómez, A.2    Machado, A.3    Ayala, A.4
  • 62
    • 72649102374 scopus 로고    scopus 로고
    • Protection against reactive oxygen species by selenoproteins
    • 2-s2.0-72649102374 10.1016/j.bbagen.2009.02.014
    • Steinbrenner H., Sies H., Protection against reactive oxygen species by selenoproteins. Biochimica et Biophysica Acta: General Subjects 2009 1790 11 1478 1485 2-s2.0-72649102374 10.1016/j.bbagen.2009.02.014
    • (2009) Biochimica et Biophysica Acta: General Subjects , vol.1790 , Issue.11 , pp. 1478-1485
    • Steinbrenner, H.1    Sies, H.2
  • 63
    • 18544371009 scopus 로고    scopus 로고
    • Metals, toxicity and oxidative stress
    • DOI 10.2174/0929867053764635
    • Valko M., Morris H., Cronin M. T. D., Metals, toxicity and oxidative stress. Current Medicinal Chemistry 2005 12 10 1161 1208 2-s2.0-18544371009 10.2174/0929867053764635 (Pubitemid 40655271)
    • (2005) Current Medicinal Chemistry , vol.12 , Issue.10 , pp. 1161-1208
    • Valko, M.1    Morris, H.2    Cronin, M.T.D.3
  • 64
    • 34547673497 scopus 로고    scopus 로고
    • Peroxynitrite: Biochemistry, pathophysiology and development of therapeutics
    • DOI 10.1038/nrd2222, PII NRD2222
    • Szabó C., Ischiropoulos H., Radi R., Peroxynitrite: biochemistry, pathophysiology and development of therapeutics. Nature Reviews Drug Discovery 2007 6 8 662 680 (Pubitemid 47207751)
    • (2007) Nature Reviews Drug Discovery , vol.6 , Issue.8 , pp. 662-680
    • Szabo, C.1    Ischiropoulos, H.2    Radi, R.3
  • 65
    • 0037184780 scopus 로고    scopus 로고
    • Biological reactivity and biomarkers of the neutrophil oxidant, hypochlorous acid
    • DOI 10.1016/S0300-483X(02)00286-X, PII S0300483X0200286X
    • Winterbourn C. C., Biological reactivity and biomarkers of the neutrophil oxidant, hypochlorous acid. Toxicology 2002 181-182 223 227 2-s2.0-0037184780 10.1016/S0300-483X(02)00286-X (Pubitemid 36015703)
    • (2002) Toxicology , vol.181-182 , pp. 223-227
    • Winterbourn, C.C.1
  • 66
    • 33646826643 scopus 로고    scopus 로고
    • Modification of low-density lipoprotein by myeloperoxidase-derived oxidants and reagent hypochlorous acid
    • DOI 10.1016/j.bbalip.2006.03.024, PII S1388198106000990
    • Malle E., Marsche G., Arnhold J., Davies M. J., Modification of low-density lipoprotein by myeloperoxidase-derived oxidants and reagent hypochlorous acid. Biochimica et Biophysica Acta: Molecular and Cell Biology of Lipids 2006 1761 4 392 415 2-s2.0-33646826643 10.1016/j.bbalip.2006.03.024 (Pubitemid 43776460)
    • (2006) Biochimica et Biophysica Acta - Molecular and Cell Biology of Lipids , vol.1761 , Issue.4 , pp. 392-415
    • Malle, E.1    Marsche, G.2    Arnhold, J.3    Davies, M.J.4
  • 69
    • 67349228432 scopus 로고    scopus 로고
    • Singlet-oxygen-mediated amino acid and protein oxidation: Formation of tryptophan peroxides and decomposition products
    • 2-s2.0-67349228432 10.1016/j.freeradbiomed.2009.04.015
    • Gracanin M., Hawkins C. L., Pattison D. I., Davies M. J., Singlet-oxygen-mediated amino acid and protein oxidation: formation of tryptophan peroxides and decomposition products. Free Radical Biology and Medicine 2009 47 1 92 102 2-s2.0-67349228432 10.1016/j.freeradbiomed.2009.04.015
    • (2009) Free Radical Biology and Medicine , vol.47 , Issue.1 , pp. 92-102
    • Gracanin, M.1    Hawkins, C.L.2    Pattison, D.I.3    Davies, M.J.4
  • 70
    • 0037564169 scopus 로고    scopus 로고
    • Singlet oxygen-mediated damage to proteins and its consequences
    • DOI 10.1016/S0006-291X(03)00817-9
    • Davies M. J., Singlet oxygen-mediated damage to proteins and its consequences. Biochemical and Biophysical Research Communications 2003 305 3 761 770 (Pubitemid 36579294)
    • (2003) Biochemical and Biophysical Research Communications , vol.305 , Issue.3 , pp. 761-770
    • Davies, M.J.1
  • 72
    • 37849013375 scopus 로고    scopus 로고
    • Advanced lipid peroxidation end products in oxidative damage to proteins. Potential role in diseases and therapeutic prospects for the inhibitors
    • 2-s2.0-37849013375 10.1038/sj.bjp.0707395
    • Negre-Salvayre A., Coatrieux C., Ingueneau C., Salvayre R., Advanced lipid peroxidation end products in oxidative damage to proteins. Potential role in diseases and therapeutic prospects for the inhibitors. British Journal of Pharmacology 2008 153 1 6 20 2-s2.0-37849013375 10.1038/sj.bjp.0707395
    • (2008) British Journal of Pharmacology , vol.153 , Issue.1 , pp. 6-20
    • Negre-Salvayre, A.1    Coatrieux, C.2    Ingueneau, C.3    Salvayre, R.4
  • 73
    • 84884540572 scopus 로고    scopus 로고
    • Malondialdehyde regulates glucose-stimulated insulin secretion in murine islets via TCF7L2-dependent Wnt signaling pathway
    • Wang X., Lei X. G., Wang J., Malondialdehyde regulates glucose-stimulated insulin secretion in murine islets via TCF7L2-dependent Wnt signaling pathway. Molecular and Cellular Endocrinology 2014 382 1 8 16
    • (2014) Molecular and Cellular Endocrinology , vol.382 , Issue.1 , pp. 8-16
    • Wang, X.1    Lei, X.G.2    Wang, J.3
  • 74
    • 0037163121 scopus 로고    scopus 로고
    • Sp1 and Sp3 transcription factors mediate malondialdehyde-induced collagen α1(I) gene expression in cultured hepatic stellate cells
    • DOI 10.1074/jbc.M203368200
    • García-Ruiz I., de la Torre P., Díaz T., Esteban E., Fernández I., Muñoz-Yagüe T., Solís-Herruzo J. A., Sp1 and Sp3 transcription factors mediate malondialdehyde-induced collagen alpha 1(I) gene expression in cultured hepatic stellate cells. The Journal of Biological Chemistry 2002 277 34 30551 30558 (Pubitemid 34970748)
    • (2002) Journal of Biological Chemistry , vol.277 , Issue.34 , pp. 30551-30558
    • Garcia-Ruiz, I.1    De La Torre, P.2    Diaz, T.3    Esteban, E.4    Fernandez, I.5    Munoz-Yague, T.6    Solis-Herruzo, J.A.7
  • 75
    • 77956611413 scopus 로고    scopus 로고
    • The role of Sp1 and Sp3 in normal and cancer cell biology
    • Li L., Davie J. R., The role of Sp1 and Sp3 in normal and cancer cell biology. Annals of Anatomy: Anatomischer Anzeiger 2010 192 5 275 283
    • (2010) Annals of Anatomy: Anatomischer Anzeiger , vol.192 , Issue.5 , pp. 275-283
    • Li, L.1    Davie, J.R.2
  • 77
    • 47049109950 scopus 로고    scopus 로고
    • DNA adducts with lipid peroxidation products
    • 2-s2.0-47049109950 10.1074/jbc.R700051200
    • Blair I. A., DNA adducts with lipid peroxidation products. Journal of Biological Chemistry 2008 283 23 15545 15549 2-s2.0-47049109950 10.1074/jbc.R700051200
    • (2008) Journal of Biological Chemistry , vol.283 , Issue.23 , pp. 15545-15549
    • Blair, I.A.1
  • 80
    • 84863460571 scopus 로고    scopus 로고
    • Association between age-related decline of kidney function and plasma malondialdehyde
    • Li G., Chen Y., Hu H., Liu L., Hu X., Wang J., Shi W., Yin D., Association between age-related decline of kidney function and plasma malondialdehyde. Rejuvenation Research 2012 15 3 257 264
    • (2012) Rejuvenation Research , vol.15 , Issue.3 , pp. 257-264
    • Li, G.1    Chen, Y.2    Hu, H.3    Liu, L.4    Hu, X.5    Wang, J.6    Shi, W.7    Yin, D.8
  • 82
    • 42449119729 scopus 로고    scopus 로고
    • Proinflammatory effects of advanced lipoxidation end products in monocytes
    • 2-s2.0-42449119729 10.2337/db07-1204
    • Shanmugam N., Figarola J. L., Li Y., Swiderski P. M., Rahbar S., Natarajan R., Proinflammatory effects of advanced lipoxidation end products in monocytes. Diabetes 2008 57 4 879 888 2-s2.0-42449119729 10.2337/db07-1204
    • (2008) Diabetes , vol.57 , Issue.4 , pp. 879-888
    • Shanmugam, N.1    Figarola, J.L.2    Li, Y.3    Swiderski, P.M.4    Rahbar, S.5    Natarajan, R.6
  • 83
    • 33745995174 scopus 로고    scopus 로고
    • Investigation of protein oxidation and lipid peroxidation in patients with rheumatoid arthritis
    • DOI 10.1002/cbf.1257
    • Baskol G., Demir H., Baskol M., Kilic E., Ates F., Karakukcu C., Ustdal M., Investigation of protein oxidation and lipid peroxidation in patients with rheumatoid arthritis. Cell Biochemistry and Function 2006 24 4 307 311 2-s2.0-33745995174 10.1002/cbf.1257 (Pubitemid 44065233)
    • (2006) Cell Biochemistry and Function , vol.24 , Issue.4 , pp. 307-311
    • Baskol, G.1    Demir, H.2    Baskol, M.3    Kilic, E.4    Ates, F.5    Karakukcu, C.6    Ustdal, M.7
  • 86
    • 0024494718 scopus 로고
    • On the mechanism of prostacyclin and thromboxane A2 biosynthesis
    • 2-s2.0-0024494718
    • Hecker M., Ullrich V., On the mechanism of prostacyclin and thromboxane A2 biosynthesis. Journal of Biological Chemistry 1989 264 1 141 150 2-s2.0-0024494718
    • (1989) Journal of Biological Chemistry , vol.264 , Issue.1 , pp. 141-150
    • Hecker, M.1    Ullrich, V.2
  • 88
    • 84867576603 scopus 로고    scopus 로고
    • Glutathione promotes prostaglandin H synthase (cyclooxygenase)-dependent formation of malondialdehyde and 15(S)-8-iso-prostaglandin F2 α
    • Tsikas D., Suchy M. T., Niemann J., Tossios P., Schneider Y., Rothmann S., Gutzki F. M., Frölich J. C., Stichtenoth D. O., Glutathione promotes prostaglandin H synthase (cyclooxygenase)-dependent formation of malondialdehyde and 15(S)-8-iso-prostaglandin F2 α FEBS Letters 2012 586 20 3723 3730
    • (2012) FEBS Letters , vol.586 , Issue.20 , pp. 3723-3730
    • Tsikas, D.1    Suchy, M.T.2    Niemann, J.3    Tossios, P.4    Schneider, Y.5    Rothmann, S.6    Gutzki, F.M.7    Frölich, J.C.8    Stichtenoth, D.O.9
  • 89
    • 70849134064 scopus 로고    scopus 로고
    • Convergence of the 5-LOX and COX-2 pathways: Heme-catalyzed cleavage of the 5S-HETE-derived di-endoperoxide into aldehyde fragments
    • 2-s2.0-70849134064 10.1194/jlr.M900181-JLR200
    • Griesser M., Boeglin W. E., Suzuki T., Schneider C., Convergence of the 5-LOX and COX-2 pathways: heme-catalyzed cleavage of the 5S-HETE-derived di-endoperoxide into aldehyde fragments. Journal of Lipid Research 2009 50 12 2455 2462 2-s2.0-70849134064 10.1194/jlr.M900181-JLR200
    • (2009) Journal of Lipid Research , vol.50 , Issue.12 , pp. 2455-2462
    • Griesser, M.1    Boeglin, W.E.2    Suzuki, T.3    Schneider, C.4
  • 92
    • 83855164044 scopus 로고    scopus 로고
    • The thromboxane synthase and receptor signaling pathway in cancer: An emerging paradigm in cancer progression and metastasis
    • 2-s2.0-83855164044 10.1007/s10555-011-9297-9
    • Ekambaram P., Lambiv W., Cazzolli R., Ashton A. W., Honn K. V., The thromboxane synthase and receptor signaling pathway in cancer: an emerging paradigm in cancer progression and metastasis. Cancer and Metastasis Reviews 2011 30 3-4 397 408 2-s2.0-83855164044 10.1007/s10555-011-9297-9
    • (2011) Cancer and Metastasis Reviews , vol.30 , Issue.3-4 , pp. 397-408
    • Ekambaram, P.1    Lambiv, W.2    Cazzolli, R.3    Ashton, A.W.4    Honn, K.V.5
  • 93
    • 47249129136 scopus 로고    scopus 로고
    • Cyclooxygenase-2 in synaptic signaling
    • DOI 10.2174/138161208784480144
    • Yang H., Chen C., Cyclooxygenase-2 in synaptic signaling. Current Pharmaceutical Design 2008 14 14 1443 1451 (Pubitemid 351993045)
    • (2008) Current Pharmaceutical Design , vol.14 , Issue.14 , pp. 1443-1451
    • Yang, H.1    Chen, C.2
  • 94
    • 0017070586 scopus 로고
    • Autoxidation of polyunsaturated fatty acids: II. A suggested mechanism for the formation of TBA reactive materials from prostaglandin like endoperoxides
    • 2-s2.0-0017070586
    • Pryor W. A., Stanley J. P., Blair E., Autoxidation of polyunsaturated fatty acids: II. A suggested mechanism for the formation of TBA reactive materials from prostaglandin like endoperoxides. Lipids 1976 11 5 370 379 2-s2.0-0017070586
    • (1976) Lipids , vol.11 , Issue.5 , pp. 370-379
    • Pryor, W.A.1    Stanley, J.P.2    Blair, E.3
  • 95
    • 47049114843 scopus 로고    scopus 로고
    • Human biochemistry of the isoprostane pathway
    • 2-s2.0-47049114843 10.1074/jbc.R700047200
    • Milne G. L., Yin H., Morrow J. D., Human biochemistry of the isoprostane pathway. Journal of Biological Chemistry 2008 283 23 15533 15537 2-s2.0-47049114843 10.1074/jbc.R700047200
    • (2008) Journal of Biological Chemistry , vol.283 , Issue.23 , pp. 15533-15537
    • Milne, G.L.1    Yin, H.2    Morrow, J.D.3
  • 96
  • 97
    • 33846963508 scopus 로고    scopus 로고
    • 2α is generated from the isoprostane pathway and not the cyclooxygenase in humans
    • DOI 10.1074/jbc.M608975200
    • Yin H., Gao L., Tai H.-H., Murphey L. J., Porter N. A., Morrow J. D., Urinary prostaglandin F2 α is generated from the isoprostane pathway and not the cyclooxygenase in humans. Journal of Biological Chemistry 2007 282 1 329 336 2-s2.0-33846963508 10.1074/jbc.M608975200 (Pubitemid 47076631)
    • (2007) Journal of Biological Chemistry , vol.282 , Issue.1 , pp. 329-336
    • Yin, H.1    Gao, L.2    Tai, H.-H.3    Murphey, L.J.4    Porter, N.A.5    Morrow, J.D.6
  • 98
    • 45549090724 scopus 로고    scopus 로고
    • Formation of highly reactive cyclopentenone isoprostane compounds (A 3/J3-isoprostanes) in vivo from eicosapentaenoic acid
    • 2-s2.0-45549090724 10.1074/jbc.M800122200
    • Brooks J. D., Milne G. L., Yin H., Sanchez S. C., Porter N. A., Morrow J. D., Formation of highly reactive cyclopentenone isoprostane compounds (A 3/J3-isoprostanes) in vivo from eicosapentaenoic acid. Journal of Biological Chemistry 2008 283 18 12043 12055 2-s2.0-45549090724 10.1074/jbc.M800122200
    • (2008) Journal of Biological Chemistry , vol.283 , Issue.18 , pp. 12043-12055
    • Brooks, J.D.1    Milne, G.L.2    Yin, H.3    Sanchez, S.C.4    Porter, N.A.5    Morrow, J.D.6
  • 99
    • 17444403972 scopus 로고    scopus 로고
    • The biochemistry of the isoprostane, neuroprostane, and isofuran pathways of lipid peroxidation
    • Roberts L. J. II, Fessel J. P., Davies S. S., The biochemistry of the isoprostane, neuroprostane, and isofuran pathways of lipid peroxidation. Brain Pathology 2005 15 2 143 148 2-s2.0-17444403972 (Pubitemid 40546170)
    • (2005) Brain Pathology , vol.15 , Issue.2 , pp. 143-148
    • Roberts II, L.J.1    Fessel, J.P.2    Davies, S.S.3
  • 100
    • 77957754592 scopus 로고    scopus 로고
    • New hypotheses on the pathways of formation of malondialdehyde and isofurans
    • 2-s2.0-77957754592 10.1016/j.freeradbiomed.2010.08.012
    • Onyango A. N., Baba N., New hypotheses on the pathways of formation of malondialdehyde and isofurans. Free Radical Biology and Medicine 2010 49 10 1594 1600 2-s2.0-77957754592 10.1016/j.freeradbiomed.2010.08.012
    • (2010) Free Radical Biology and Medicine , vol.49 , Issue.10 , pp. 1594-1600
    • Onyango, A.N.1    Baba, N.2
  • 101
    • 0019920815 scopus 로고
    • Metabolism of malonaldehyde in vivo and in vitro
    • DOI 10.1007/BF02535193
    • Siu G. M., Draper H. H., Metabolism of malonaldehyde in vivo and in vitro. Lipids 1982 17 5 349 355 2-s2.0-0019920815 (Pubitemid 12058259)
    • (1982) Lipids , vol.17 , Issue.5 , pp. 349-355
    • Siu, G.M.1    Draper, H.H.2
  • 102
    • 0022354502 scopus 로고
    • Distribution and oxidation of malondialdehyde in mice
    • DOI 10.1016/0090-6980(85)90188-1
    • Marnett L. J., Buck J., Tuttle M. A., Basu A. K., Bull A. W., Distribution and oxidation of malondialdehyde in mice. Prostaglandins 1985 30 2 241 254 2-s2.0-0022354502 (Pubitemid 16245431)
    • (1985) Prostaglandins , vol.30 , Issue.2 , pp. 241-254
    • Marnett, L.J.1    Buck, J.2    Tuttle, M.A.3
  • 103
    • 31544481909 scopus 로고    scopus 로고
    • A new role of phosphoglucose isomerase. Involvement of the glycolytic enzyme in aldehyde metabolism
    • DOI 10.1007/s10541-005-0255-4
    • Agadjanyan Z. S., Dmitriev L. F., Dugin S. F., A new role of phosphoglucose isomerase. Involvement of the glycolytic enzyme in aldehyde metabolism. Biochemistry 2005 70 11 1251 1255 2-s2.0-29144484253 10.1007/s10541-005-0255-4 (Pubitemid 41802121)
    • (2005) Biochemistry (Moscow) , vol.70 , Issue.11 , pp. 1251-1255
    • Agadjanyan, Z.S.1    Dmitriev, L.F.2    Dugin, S.F.3
  • 105
  • 106
    • 69049109744 scopus 로고    scopus 로고
    • Biomarkers of oxidative and nitrosative damage in Alzheimer's disease and mild cognitive impairment
    • 2-s2.0-69049109744 10.1016/j.arr.2009.04.002
    • Mangialasche F., Polidori M. C., Monastero R., Ercolani S., Camarda C., Cecchetti R., Mecocci P., Biomarkers of oxidative and nitrosative damage in Alzheimer's disease and mild cognitive impairment. Ageing Research Reviews 2009 8 4 285 305 2-s2.0-69049109744 10.1016/j.arr.2009.04.002
    • (2009) Ageing Research Reviews , vol.8 , Issue.4 , pp. 285-305
    • Mangialasche, F.1    Polidori, M.C.2    Monastero, R.3    Ercolani, S.4    Camarda, C.5    Cecchetti, R.6    Mecocci, P.7
  • 107
    • 20444373701 scopus 로고    scopus 로고
    • Proteins in human brain cortex are modified by oxidation, glycoxidation, and lipoxidation: Effects of Alzheimer disease and identification of lipoxidation targets
    • DOI 10.1074/jbc.M502255200
    • Pamplona R., Dalfó E., Ayala V., Bellmunt M. J., Prat J., Ferrer I., Portero-Otín M., Proteins in human brain cortex are modified by oxidation, glycoxidation, and lipoxidation: effects of Alzheimer disease and identification of lipoxidation targets. Journal of Biological Chemistry 2005 280 22 21522 21530 2-s2.0-20444373701 10.1074/jbc.M502255200 (Pubitemid 40805718)
    • (2005) Journal of Biological Chemistry , vol.280 , Issue.22 , pp. 21522-21530
    • Pamplona, R.1    Dalfo, E.2    Ayala, V.3    Bellmunt, M.J.4    Prat, J.5    Ferrer, I.6    Portero-Otin, M.7
  • 108
    • 84857037658 scopus 로고    scopus 로고
    • Peripheral markers in neurodegenerative patients and their first-degree relatives
    • 2-s2.0-84857037658 10.1016/j.jns.2011.11.001
    • Cristalli D. O., Arnal N., Marra F. A., De Alaniz M. J. T., Marra C. A., Peripheral markers in neurodegenerative patients and their first-degree relatives. Journal of the Neurological Sciences 2012 314 1-2 48 56 2-s2.0-84857037658 10.1016/j.jns.2011.11.001
    • (2012) Journal of the Neurological Sciences , vol.314 , Issue.1-2 , pp. 48-56
    • Cristalli, D.O.1    Arnal, N.2    Marra, F.A.3    De Alaniz, M.J.T.4    Marra, C.A.5
  • 109
    • 33749986298 scopus 로고    scopus 로고
    • Free radicals and antioxidants in normal physiological functions and human disease
    • DOI 10.1016/j.biocel.2006.07.001, PII S1357272506002196
    • Valko M., Leibfritz D., Moncol J., Cronin M. T. D., Mazur M., Telser J., Free radicals and antioxidants in normal physiological functions and human disease. International Journal of Biochemistry and Cell Biology 2007 39 1 44 84 2-s2.0-33749986298 10.1016/j.biocel.2006.07.001 (Pubitemid 44566469)
    • (2007) International Journal of Biochemistry and Cell Biology , vol.39 , Issue.1 , pp. 44-84
    • Valko, M.1    Leibfritz, D.2    Moncol, J.3    Cronin, M.T.D.4    Mazur, M.5    Telser, J.6
  • 110
    • 84872471697 scopus 로고    scopus 로고
    • Oxidative stress in Alzheimer's disease and mild cognitive impairment with high sensitivity and specificity
    • López N., Tormo C., De Blas I., Llinares I., Alom J., Oxidative stress in Alzheimer's disease and mild cognitive impairment with high sensitivity and specificity. Journal of Alzheimer's Disease 2013 33 3 823 829
    • (2013) Journal of Alzheimer's Disease , vol.33 , Issue.3 , pp. 823-829
    • López, N.1    Tormo, C.2    De Blas, I.3    Llinares, I.4    Alom, J.5
  • 112
    • 0036970014 scopus 로고    scopus 로고
    • Plasma susceptibility to free radical-induced antioxidant consumption and lipid peroxidation is increased in very old subjects with Alzheimer disease
    • Polidori M. C., Mecocci P., Plasma susceptibility to free radical-induced antioxidant consumption and lipid peroxidation is increased in very old subjects with Alzheimer disease. Journal of Alzheimer's Disease 2002 4 6 517 522 2-s2.0-0036970014 (Pubitemid 36142841)
    • (2002) Journal of Alzheimer's Disease , vol.4 , Issue.6 , pp. 517-522
    • Polidori, M.C.1    Mecocci, P.2
  • 113
    • 72649096229 scopus 로고    scopus 로고
    • Changes of some oxidative stress markers in the serum of patients with mild cognitive impairment and Alzheimer's disease
    • 2-s2.0-72649096229 10.1016/j.neulet.2009.11.033
    • Padurariu M., Ciobica A., Hritcu L., Stoica B., Bild W., Stefanescu C., Changes of some oxidative stress markers in the serum of patients with mild cognitive impairment and Alzheimer's disease. Neuroscience Letters 2010 469 1 6 10 2-s2.0-72649096229 10.1016/j.neulet.2009.11.033
    • (2010) Neuroscience Letters , vol.469 , Issue.1 , pp. 6-10
    • Padurariu, M.1    Ciobica, A.2    Hritcu, L.3    Stoica, B.4    Bild, W.5    Stefanescu, C.6
  • 114
    • 84884902071 scopus 로고    scopus 로고
    • Oxidative damage to macromolecules in human Parkinson disease and the rotenone model
    • Sanders L. H., Timothy Greenamyre J., Oxidative damage to macromolecules in human Parkinson disease and the rotenone model. Free Radical Biology and Medicine 2013 62 111 120
    • (2013) Free Radical Biology and Medicine , vol.62 , pp. 111-120
    • Sanders, L.H.1    Timothy Greenamyre, J.2
  • 115
    • 79961207643 scopus 로고    scopus 로고
    • Evaluation of Markers of oxidative stress, antioxidant function and astrocytic proliferation in the striatum and frontal cortex of Parkinson's disease brains
    • 2-s2.0-79961207643 10.1007/s11064-011-0471-9
    • Mythri R. B., Venkateshappa C., Harish G., Mahadevan A., Muthane U. B., Yasha T. C., Srinivas Bharath M. M., Shankar S. K., Evaluation of Markers of oxidative stress, antioxidant function and astrocytic proliferation in the striatum and frontal cortex of Parkinson's disease brains. Neurochemical Research 2011 36 8 1452 1463 2-s2.0-79961207643 10.1007/s11064-011-0471-9
    • (2011) Neurochemical Research , vol.36 , Issue.8 , pp. 1452-1463
    • Mythri, R.B.1    Venkateshappa, C.2    Harish, G.3    Mahadevan, A.4    Muthane, U.B.5    Yasha, T.C.6    Srinivas Bharath, M.M.7    Shankar, S.K.8
  • 117
    • 0023889360 scopus 로고
    • Increased erythrocyte susceptibility to lipid peroxidation in human Parkinson's disease
    • Kilinç A., Yalçin A. S., Yalçin D., Taga Y., Emerk K., Increased erythrocyte susceptibility to lipid peroxidation in human Parkinson's disease. Neuroscience Letters 1988 87 3 307 310
    • (1988) Neuroscience Letters , vol.87 , Issue.3 , pp. 307-310
    • Kilinç, A.1    Yalçin, A.S.2    Yalçin, D.3    Taga, Y.4    Emerk, K.5
  • 118
    • 77957562993 scopus 로고    scopus 로고
    • The role of oxidative stress in amyotrophic lateral sclerosis and Parkinson's disease
    • 2-s2.0-77957562993 10.1007/s11064-010-0212-5
    • Baillet A., Chanteperdrix V., Trocmé C., Casez P., Garrel C., Besson G., The role of oxidative stress in amyotrophic lateral sclerosis and Parkinson's disease. Neurochemical Research 2010 35 10 1530 1537 2-s2.0-77957562993 10.1007/s11064-010-0212-5
    • (2010) Neurochemical Research , vol.35 , Issue.10 , pp. 1530-1537
    • Baillet, A.1    Chanteperdrix, V.2    Trocmé, C.3    Casez, P.4    Garrel, C.5    Besson, G.6
  • 119
    • 60849129435 scopus 로고    scopus 로고
    • Increased oxidative damage in peripheral blood correlates with severity of Parkinson's disease
    • Chen C. M., Liu J. L., Wu Y. R., Chen Y. C., Cheng H. S., Cheng M. L., Chiu D. T. Y., Increased oxidative damage in peripheral blood correlates with severity of Parkinson's disease. Neurobiology of Disease 2009 33 3 429 435
    • (2009) Neurobiology of Disease , vol.33 , Issue.3 , pp. 429-435
    • Chen, C.M.1    Liu, J.L.2    Wu, Y.R.3    Chen, Y.C.4    Cheng, H.S.5    Cheng, M.L.6    Chiu, D.T.Y.7
  • 120
    • 0026728880 scopus 로고
    • Oxygen free radical producing activity of polymorphonuclear leukocytes in patients with Parkinson's disease
    • 2-s2.0-0026728880
    • Kalra J., Rajput A. H., Mantha S. V., Chaudhary A. K., Prasad K., Oxygen free radical producing activity of polymorphonuclear leukocytes in patients with Parkinson's disease. Molecular and Cellular Biochemistry 1992 112 2 181 186 2-s2.0-0026728880
    • (1992) Molecular and Cellular Biochemistry , vol.112 , Issue.2 , pp. 181-186
    • Kalra, J.1    Rajput, A.H.2    Mantha, S.V.3    Chaudhary, A.K.4    Prasad, K.5
  • 121
    • 34547788327 scopus 로고    scopus 로고
    • Peripheral blood markers of oxidative stress in Parkinson's disease
    • DOI 10.1159/000103641
    • Younes-Mhenni S., Frih-Ayed M., Kerkeni A., Bost M., Chazot G., Peripheral blood markers of oxidative stress in Parkinson's disease. European Neurology 2007 58 2 78 83 2-s2.0-34547788327 10.1159/000103641 (Pubitemid 47237650)
    • (2007) European Neurology , vol.58 , Issue.2 , pp. 78-83
    • Younes-Mhenni, S.1    Frih-Ayed, M.2    Kerkeni, A.3    Bost, M.4    Chazot, G.5
  • 122
    • 0041731614 scopus 로고    scopus 로고
    • Malondialdehyde, a product of lipid peroxidation, is mutagenic in human cells
    • DOI 10.1074/jbc.M212549200
    • Niedernhofer L. J., Daniels J. S., Rouzer C. A., Greene R. E., Marnett L. J., Malondialdehyde, a product of lipid peroxidation, is mutagenic in human cells. Journal of Biological Chemistry 2003 278 33 31426 31433 2-s2.0-0041731614 10.1074/jbc.M212549200 (Pubitemid 36994664)
    • (2003) Journal of Biological Chemistry , vol.278 , Issue.33 , pp. 31426-31433
    • Niedernhofer, L.J.1    Daniels, J.S.2    Rouzer, C.A.3    Greene, R.E.4    Marnett, L.J.5
  • 123
    • 23944478990 scopus 로고    scopus 로고
    • A review of recent studies on malondialdehyde as toxic molecule and biological marker of oxidative stress
    • DOI 10.1016/j.numecd.2005.05.003
    • Del Rio D., Stewart A. J., Pellegrini N., A review of recent studies on malondialdehyde as toxic molecule and biological marker of oxidative stress. Nutrition, Metabolism and Cardiovascular Diseases 2005 15 4 316 328 2-s2.0-23944478990 10.1016/j.numecd.2005.05.003 (Pubitemid 41190990)
    • (2005) Nutrition, Metabolism and Cardiovascular Diseases , vol.15 , Issue.4 , pp. 316-328
    • Del Rio, D.1    Stewart, A.J.2    Pellegrini, N.3
  • 124
    • 0344630253 scopus 로고    scopus 로고
    • Induction of frameshift and base pair substitution mutations by the major DNA adduct of the endogenous carcinogen malondialdehyde
    • DOI 10.1073/pnas.2332176100
    • VanderVeen L. A., Hashim M. F., Shyr Y., Marnett L. J., Induction of frameshift and base pair substitution mutations by the major DNA adduct of the endogenous carcinogen malondialdehyde. Proceedings of the National Academy of Sciences of the United States of America 2003 100 24 14247 14252 2-s2.0-0344630253 10.1073/pnas.2332176100 (Pubitemid 37499222)
    • (2003) Proceedings of the National Academy of Sciences of the United States of America , vol.100 , Issue.SUPPL. 2 , pp. 14247-14252
    • VanderVeen, L.A.1    Hashim, M.F.2    Shyr, Y.3    Marnett, L.J.4
  • 126
    • 84857055180 scopus 로고    scopus 로고
    • Role of polyunsaturated fatty acids and lipid peroxidation on colorectal cancer risk and treatments
    • 2-s2.0-84857055180 10.1097/MCO.0b013e32834feab4
    • Cai F., Dupertuis Y. M., Pichard C., Role of polyunsaturated fatty acids and lipid peroxidation on colorectal cancer risk and treatments. Current Opinion in Clinical Nutrition and Metabolic Care 2012 15 2 99 106 2-s2.0-84857055180 10.1097/MCO.0b013e32834feab4
    • (2012) Current Opinion in Clinical Nutrition and Metabolic Care , vol.15 , Issue.2 , pp. 99-106
    • Cai, F.1    Dupertuis, Y.M.2    Pichard, C.3
  • 127
    • 34548508693 scopus 로고    scopus 로고
    • Lipid peroxidation-induced DNA damage in cancer-prone inflammatory diseases: A review of published adduct types and levels in humans
    • DOI 10.1016/j.freeradbiomed.2007.07.012, PII S0891584907004935
    • Nair U., Bartsch H., Nair J., Lipid peroxidation-induced DNA damage in cancer-prone inflammatory diseases: a review of published adduct types and levels in humans. Free Radical Biology and Medicine 2007 43 8 1109 1120 2-s2.0-34548508693 10.1016/j.freeradbiomed.2007.07.012 (Pubitemid 47374441)
    • (2007) Free Radical Biology and Medicine , vol.43 , Issue.8 , pp. 1109-1120
    • Nair, U.1    Bartsch, H.2    Nair, J.3
  • 128
    • 27844525474 scopus 로고    scopus 로고
    • Accumulation of lipid peroxidation-derived DNA lesions: Potential lead markers for chemoprevention of inflammation-driven malignancies
    • DOI 10.1016/j.mrfmmm.2005.04.013, PII S0027510705002836, Mechanistic Approaches to Chemoprevention of Mutation and Cancer
    • Bartsch H., Nair J., Accumulation of lipid peroxidation-derived DNA lesions: potential lead markers for chemoprevention of inflammation-driven malignancies. Mutation Research: Fundamental and Molecular Mechanisms of Mutagenesis 2005 591 1-2 34 44 2-s2.0-27844525474 10.1016/j.mrfmmm.2005.04.013 (Pubitemid 41654838)
    • (2005) Mutation Research - Fundamental and Molecular Mechanisms of Mutagenesis , vol.591 , Issue.1-2 , pp. 34-44
    • Bartsch, H.1    Nair, J.2
  • 130
    • 84862546573 scopus 로고    scopus 로고
    • Inflammation and cancer: Chemical approaches to mechanisms, imaging, and treatment
    • Marnett L. J., Inflammation and cancer: chemical approaches to mechanisms, imaging, and treatment. Journal of Organic Chemistry 2012 77 12 5224 5238
    • (2012) Journal of Organic Chemistry , vol.77 , Issue.12 , pp. 5224-5238
    • Marnett, L.J.1
  • 131
    • 84887620267 scopus 로고    scopus 로고
    • Cell death and diseases related to oxidative stress: 4-hydroxynonenal (HNE) in the balance
    • Dalleau S., Baradat M., Guéraud F., Huc L., Cell death and diseases related to oxidative stress: 4-hydroxynonenal (HNE) in the balance. Cell Death and Differentiation 2013 20 12 1615 1630
    • (2013) Cell Death and Differentiation , vol.20 , Issue.12 , pp. 1615-1630
    • Dalleau, S.1    Baradat, M.2    Guéraud, F.3    Huc, L.4
  • 132
    • 84883890972 scopus 로고    scopus 로고
    • Oxidative stress and lipid peroxidation products in cancer progression and therapy
    • 137289 10.5402/2012/137289
    • Barrera G., Oxidative stress and lipid peroxidation products in cancer progression and therapy. ISRN Oncology 2012 2012 21 137289 10.5402/2012/137289
    • (2012) ISRN Oncology , vol.2012 , pp. 21
    • Barrera, G.1
  • 134
    • 67649933847 scopus 로고    scopus 로고
    • Chemistry and biology of DNA containing 1,N2-deoxyguanosine adducts of the α, β -unsaturated aldehydes acrolein, crotonaldehyde, and 4-hydroxynonenal
    • 2-s2.0-67649933847 10.1021/tx9000489
    • Minko I. G., Kozekov I. D., Harris T. M., Rizzo C. J., Lloyd R. S., Stone M. P., Chemistry and biology of DNA containing 1,N2-deoxyguanosine adducts of the α β -unsaturated aldehydes acrolein, crotonaldehyde, and 4-hydroxynonenal. Chemical Research in Toxicology 2009 22 5 759 778 2-s2.0-67649933847 10.1021/tx9000489
    • (2009) Chemical Research in Toxicology , vol.22 , Issue.5 , pp. 759-778
    • Minko, I.G.1    Kozekov, I.D.2    Harris, T.M.3    Rizzo, C.J.4    Lloyd, R.S.5    Stone, M.P.6
  • 136
    • 0345017021 scopus 로고    scopus 로고
    • Formation of trans-4-hydroxy-2-nonenal- and other enal-derived cyclic DNA adducts from ω-3 and ω-6 polyunsaturated fatty acids and their roles in DNA repair and human p53 gene mutation
    • DOI 10.1016/j.mrfmmm.2003.07.001
    • Chung F.-L., Pan J., Choudhury S., Roy R., Hu W., Tang M.-S., Formation of trans-4-hydroxy-2-nonenal- and other enal-derived cyclic DNA adducts from ω -3 and ω -6 polyunsaturated fatty acids and their roles in DNA repair and human p53 gene mutation. Mutation Research: Fundamental and Molecular Mechanisms of Mutagenesis 2003 531 1-2 25 36 2-s2.0-0345017021 10.1016/j.mrfmmm.2003.07.001 (Pubitemid 37456722)
    • (2003) Mutation Research - Fundamental and Molecular Mechanisms of Mutagenesis , vol.531 , Issue.1-2 , pp. 25-36
    • Chung, F.-L.1    Pan, J.2    Choudhury, S.3    Roy, R.4    Hu, W.5    Tang, M.-S.6
  • 137
    • 84860718879 scopus 로고    scopus 로고
    • Evaluating oxidative stress in human cardiovascular disease: Methodological aspects and considerations
    • Lee R., Margaritis M., Channon K. M., Antoniades C., Evaluating oxidative stress in human cardiovascular disease: methodological aspects and considerations. Current Medicinal Chemistry 2012 19 16 2504 2520
    • (2012) Current Medicinal Chemistry , vol.19 , Issue.16 , pp. 2504-2520
    • Lee, R.1    Margaritis, M.2    Channon, K.M.3    Antoniades, C.4
  • 138
    • 0034659147 scopus 로고    scopus 로고
    • Role of reactive aldehyde in cardiovascular diseases
    • DOI 10.1016/S0891-5849(00)00226-4, PII S0891584900002264
    • Uchida K., Role of reactive aldehyde in cardiovascular diseases. Free Radical Biology and Medicine 2000 28 12 1685 1696 2-s2.0-0034659147 10.1016/S0891-5849(00)00226-4 (Pubitemid 30628655)
    • (2000) Free Radical Biology and Medicine , vol.28 , Issue.12 , pp. 1685-1696
    • Uchida, K.1
  • 139
    • 84856464242 scopus 로고    scopus 로고
    • Mitochondria as a source and target of lipid peroxidation products in healthy and diseased heart
    • 2-s2.0-84856464242 10.1111/j.1440-1681.2011.05641.x
    • Anderson E. J., Katunga L. A., Willis M. S., Mitochondria as a source and target of lipid peroxidation products in healthy and diseased heart. Clinical and Experimental Pharmacology and Physiology 2012 39 2 179 193 2-s2.0-84856464242 10.1111/j.1440-1681.2011.05641.x
    • (2012) Clinical and Experimental Pharmacology and Physiology , vol.39 , Issue.2 , pp. 179-193
    • Anderson, E.J.1    Katunga, L.A.2    Willis, M.S.3
  • 140
    • 34250784999 scopus 로고    scopus 로고
    • Erythrocyte oxidative stress in clinical management of diabetes and its cardiovascular complications
    • Nwose E. U., Jelinek H. F., Richards R. S., Kerr R. G., Erythrocyte oxidative stress in clinical management of diabetes and its cardiovascular complications. British Journal of Biomedical Science 2007 64 1 35 43 2-s2.0-34250784999 (Pubitemid 46952971)
    • (2007) British Journal of Biomedical Science , vol.64 , Issue.1 , pp. 35-43
    • Nwose, E.U.1    Jelinek, H.F.2    Richards, R.S.3    Kerr, R.G.4
  • 141
    • 84887034371 scopus 로고    scopus 로고
    • Biological markers of oxidative stress: Applications to cardiovascular research and practice
    • Ho E., Karimi Galougahi K., Liu C. C., Bhindi R., Figtree G. A., Biological markers of oxidative stress: applications to cardiovascular research and practice. Redox Biology 2013 1 1 483 491
    • (2013) Redox Biology , vol.1 , Issue.1 , pp. 483-491
    • Ho, E.1    Karimi Galougahi, K.2    Liu, C.C.3    Bhindi, R.4    Figtree, G.A.5
  • 142
    • 84880251695 scopus 로고    scopus 로고
    • Effects of 4-hydroxynonenal on vascular endothelial and smooth muscle cell redox signaling and function in health and disease
    • Chapple S. J., Cheng X., Mann G. E., Effects of 4-hydroxynonenal on vascular endothelial and smooth muscle cell redox signaling and function in health and disease. Redox Biology 2013 1 1 319 331
    • (2013) Redox Biology , vol.1 , Issue.1 , pp. 319-331
    • Chapple, S.J.1    Cheng, X.2    Mann, G.E.3
  • 143
    • 70349236876 scopus 로고    scopus 로고
    • Roles of the lipid peroxidation product 4-hydroxynonenal in obesity, the metabolic syndrome, and associated vascular and neurodegenerative disorders
    • 2-s2.0-70349236876 10.1016/j.exger.2009.07.003
    • Mattson M. P., Roles of the lipid peroxidation product 4-hydroxynonenal in obesity, the metabolic syndrome, and associated vascular and neurodegenerative disorders. Experimental Gerontology 2009 44 10 625 633 2-s2.0-70349236876 10.1016/j.exger.2009.07.003
    • (2009) Experimental Gerontology , vol.44 , Issue.10 , pp. 625-633
    • Mattson, M.P.1
  • 144
    • 28444442268 scopus 로고    scopus 로고
    • 4-Hydroxynonenal and cholesterol oxidation products in atherosclerosis
    • DOI 10.1002/mnfr.200500090
    • Leonarduzzi G., Chiarpotto E., Biasi F., Poli G., 4-Hydroxynonenal and cholesterol oxidation products in atherosclerosis. Molecular Nutrition and Food Research 2005 49 11 1044 1049 2-s2.0-28444442268 10.1002/mnfr.200500090 (Pubitemid 41735564)
    • (2005) Molecular Nutrition and Food Research , vol.49 , Issue.11 , pp. 1044-1049
    • Leonarduzzi, G.1    Chiarpotto, E.2    Biasi, F.3    Poli, G.4
  • 145
    • 0034032816 scopus 로고    scopus 로고
    • The importance of lipid-derived malondialdehyde in diabetes mellitus
    • DOI 10.1007/s001250051342
    • Slatter D. A., Bolton C. H., Bailey A. J., The importance of lipid-derived malondialdehyde in diabetes mellitus. Diabetologia 2000 43 5 550 557 2-s2.0-0034032816 10.1007/s001250051342 (Pubitemid 30322071)
    • (2000) Diabetologia , vol.43 , Issue.5 , pp. 550-557
    • Slatter, D.A.1    Bolton, C.H.2    Bailey, A.J.3
  • 147
    • 77949628274 scopus 로고    scopus 로고
    • Significantly increased levels of serum malonaldehyde in type 2 diabetics with myocardial infarction
    • 2-s2.0-77949628274 10.4103/0973-3930.60006
    • Mahreen R., Mohsin M., Nasreen Z., Siraj M., Ishaq M., Significantly increased levels of serum malonaldehyde in type 2 diabetics with myocardial infarction. International Journal of Diabetes in Developing Countries 2010 30 1 49 51 2-s2.0-77949628274 10.4103/0973-3930.60006
    • (2010) International Journal of Diabetes in Developing Countries , vol.30 , Issue.1 , pp. 49-51
    • Mahreen, R.1    Mohsin, M.2    Nasreen, Z.3    Siraj, M.4    Ishaq, M.5
  • 148
    • 84901913065 scopus 로고    scopus 로고
    • Markers of oxidative stress during diabetes mellitus
    • 378790 10.1155/2013/378790
    • Tiwari B. K., Pandey K. B., Abidi A. B., Rizvi S. I., Markers of oxidative stress during diabetes mellitus. Journal of Biomarkers 2013 2013 8 378790 10.1155/2013/378790
    • (2013) Journal of Biomarkers , vol.2013 , pp. 8
    • Tiwari, B.K.1    Pandey, K.B.2    Abidi, A.B.3    Rizvi, S.I.4
  • 150
    • 84897084613 scopus 로고    scopus 로고
    • Prevention of arterial stiffening by using low-dose atorvastatin in diabetes is associated with decreased malondialdehyde
    • e90471
    • Wang C. H., Chang R. W., Ko Y. H., Tsai P. R., Wang S. S., Chen Y. S., Ko W. J., Chang C. Y., Young T. H., Chang K. C., Prevention of arterial stiffening by using low-dose atorvastatin in diabetes is associated with decreased malondialdehyde. PloS ONE 2014 9 3 e90471
    • (2014) PloS ONE , vol.9 , Issue.3
    • Wang, C.H.1    Chang, R.W.2    Ko, Y.H.3    Tsai, P.R.4    Wang, S.S.5    Chen, Y.S.6    Ko, W.J.7    Chang, C.Y.8    Young, T.H.9    Chang, K.C.10
  • 151
    • 84881230708 scopus 로고    scopus 로고
    • Reactive aldehydes - Second messengers of free radicals in diabetes mellitus
    • supplement 1
    • Jaganjac M., Tirosh O., Cohen G., Sasson S., Zarkovic N., Reactive aldehydes-second messengers of free radicals in diabetes mellitus. Free Radical Research 2013 47 supplement 1 39 48
    • (2013) Free Radical Research , vol.47 , pp. 39-48
    • Jaganjac, M.1    Tirosh, O.2    Cohen, G.3    Sasson, S.4    Zarkovic, N.5
  • 152
    • 80755152865 scopus 로고    scopus 로고
    • Role of lipid peroxidation and PPAR- δ in amplifying glucose-stimulated insulin secretion
    • 2-s2.0-80755152865 10.2337/db11-0347
    • Cohen G., Riahi Y., Shamni O., Guichardant M., Chatgilialoglu C., Ferreri C., Kaiser N., Sasson S., Role of lipid peroxidation and PPAR- δ in amplifying glucose-stimulated insulin secretion. Diabetes 2011 60 11 2830 2842 2-s2.0-80755152865 10.2337/db11-0347
    • (2011) Diabetes , vol.60 , Issue.11 , pp. 2830-2842
    • Cohen, G.1    Riahi, Y.2    Shamni, O.3    Guichardant, M.4    Chatgilialoglu, C.5    Ferreri, C.6    Kaiser, N.7    Sasson, S.8
  • 153
    • 84901915377 scopus 로고    scopus 로고
    • 4-Hydroxy-2-nonenal, an oxidative stress marker in crevicular fluid and serum in type 2 diabetes with chronic periodontitis
    • Pradeep A. R., Agarwal E., Bajaj P., Rao N. S., 4-Hydroxy-2-nonenal, an oxidative stress marker in crevicular fluid and serum in type 2 diabetes with chronic periodontitis. Contemporary Clinical Dentistry 2013 4 3 281 285
    • (2013) Contemporary Clinical Dentistry , vol.4 , Issue.3 , pp. 281-285
    • Pradeep, A.R.1    Agarwal, E.2    Bajaj, P.3    Rao, N.S.4
  • 156
    • 79955479655 scopus 로고    scopus 로고
    • PARP inhibition alleviates diabetes-induced systemic oxidative stress and neural tissue 4-hydroxynonenal adduct accumulation: Correlation with peripheral nerve function
    • 2-s2.0-79955479655 10.1016/j.freeradbiomed.2011.01.037
    • Lupachyk S., Shevalye H., Maksimchyk Y., Drel V. R., Obrosova I. G., PARP inhibition alleviates diabetes-induced systemic oxidative stress and neural tissue 4-hydroxynonenal adduct accumulation: correlation with peripheral nerve function. Free Radical Biology and Medicine 2011 50 10 1400 1409 2-s2.0-79955479655 10.1016/j.freeradbiomed.2011.01.037
    • (2011) Free Radical Biology and Medicine , vol.50 , Issue.10 , pp. 1400-1409
    • Lupachyk, S.1    Shevalye, H.2    Maksimchyk, Y.3    Drel, V.R.4    Obrosova, I.G.5
  • 157
    • 0037082350 scopus 로고    scopus 로고
    • Role of malondialdehyde-acetaldehyde adducts in liver injury
    • DOI 10.1016/S0891-5849(01)00742-0, PII S0891584901007420
    • Tuma D. J., Role of malondialdehyde-acetaldehyde adducts in liver injury. Free Radical Biology and Medicine 2002 32 4 303 308 2-s2.0-0037082350 10.1016/S0891-5849(01)00742-0 (Pubitemid 34159083)
    • (2002) Free Radical Biology and Medicine , vol.32 , Issue.4 , pp. 303-308
    • Tuma, D.J.1
  • 158
    • 0037971533 scopus 로고    scopus 로고
    • Immunohistochemical characterization of hepatic malondialdehyde and 4-hydroxynonenal modified proteins during early stages of ethanol-induced liver injury
    • Sampey B. P., Korourian S., Ronis M. J., Badger T. M., Petersen D. R., Immunohistochemical characterization of hepatic malondialdehyde and 4-hydroxynonenal modified proteins during early stages of ethanol-induced liver injury. Alcoholism: Clinical and Experimental Research 2003 27 6 1015 1022 2-s2.0-0037971533 (Pubitemid 36759392)
    • (2003) Alcoholism: Clinical and Experimental Research , vol.27 , Issue.6 , pp. 1015-1022
    • Sampey, B.P.1    Korourian, S.2    Ronis, M.J.3    Badger, T.M.4    Petersen, D.R.5
  • 159
    • 84877585734 scopus 로고    scopus 로고
    • Role of adaptive immunity in alcoholic liver disease
    • 893026 10.1155/2012/893026
    • Albano E., Role of adaptive immunity in alcoholic liver disease. International Journal of Hepatology 2012 2012 7 893026 10.1155/2012/893026
    • (2012) International Journal of Hepatology , vol.2012 , pp. 7
    • Albano, E.1
  • 160
    • 84934442823 scopus 로고    scopus 로고
    • Formation and immunological properties of aldehyde-derived protein adducts following alcohol consumption
    • 2-s2.0-43549103813 10.1007/978-1-59745-242-7-17
    • Thiele G. M., Klassen L. W., Tuma D. J., Formation and immunological properties of aldehyde-derived protein adducts following alcohol consumption. Methods in Molecular Biology 2008 447 235 257 2-s2.0-43549103813 10.1007/978-1-59745-242-7-17
    • (2008) Methods in Molecular Biology , vol.447 , pp. 235-257
    • Thiele, G.M.1    Klassen, L.W.2    Tuma, D.J.3
  • 161
    • 34250824010 scopus 로고    scopus 로고
    • Alcohol-induced oxidative stress
    • DOI 10.1016/j.lfs.2007.05.005, PII S002432050700358X
    • Das S. K., Vasudevan D. M., Alcohol-induced oxidative stress. Life Sciences 2007 81 3 177 187 2-s2.0-34250824010 10.1016/j.lfs.2007.05.005 (Pubitemid 46978376)
    • (2007) Life Sciences , vol.81 , Issue.3 , pp. 177-187
    • Das, S.K.1    Vasudevan, D.M.2
  • 162
    • 0035894063 scopus 로고    scopus 로고
    • Distribution of ethanol-induced protein adducts in vivo: Relationship to tissue injury
    • DOI 10.1016/S0891-5849(01)00744-4, PII S0891584901007444
    • Niemelä O., Distribution of ethanol-induced protein adducts in vivo: relationship to tissue injury. Free Radical Biology and Medicine 2001 31 12 1533 1538 (Pubitemid 34024975)
    • (2001) Free Radical Biology and Medicine , vol.31 , Issue.12 , pp. 1533-1538
    • Niemela, O.1
  • 164
    • 0036148631 scopus 로고    scopus 로고
    • Adduction of soluble proteins with malondialdehyde-acetaldehyde (MAA) induces antibody production and enhances T-cell proliferation
    • Willis M. S., Klassen L. W., Tuma D. J., Sorrell M. F., Thiele G. M., Adduction of soluble proteins with malondialdehyde-acetaldehyde (MAA) induces antibody production and enhances T-cell proliferation. Alcoholism: Clinical and Experimental Research 2002 26 1 94 106 2-s2.0-0036148631 (Pubitemid 34093516)
    • (2002) Alcoholism: Clinical and Experimental Research , vol.26 , Issue.1 , pp. 94-106
    • Willis, M.S.1    Klassen, L.W.2    Tuma, D.J.3    Sorrell, M.F.4    Thiele, G.M.5
  • 165
    • 84868092706 scopus 로고    scopus 로고
    • Inhibition of NF- κ B activation by 4-hydroxynonenal contributes to liver injury in a mouse model of alcoholic liver disease
    • Dou X., Li S., Wang Z., Gu D., Shen C., Yao T., Song Z., Inhibition of NF- κ B activation by 4-hydroxynonenal contributes to liver injury in a mouse model of alcoholic liver disease. The American Journal of Pathology 2012 181 5 1702 1710
    • (2012) The American Journal of Pathology , vol.181 , Issue.5 , pp. 1702-1710
    • Dou, X.1    Li, S.2    Wang, Z.3    Gu, D.4    Shen, C.5    Yao, T.6    Song, Z.7
  • 166
    • 79958058366 scopus 로고    scopus 로고
    • Overview of lipid peroxidation products and hepatic protein modification in alcoholic liver disease
    • 2-s2.0-79958058366 10.1016/j.cbi.2011.02.021
    • Smathers R. L., Galligan J. J., Stewart B. J., Petersen D. R., Overview of lipid peroxidation products and hepatic protein modification in alcoholic liver disease. Chemico-Biological Interactions 2011 192 1-2 107 112 2-s2.0-79958058366 10.1016/j.cbi.2011.02.021
    • (2011) Chemico-Biological Interactions , vol.192 , Issue.1-2 , pp. 107-112
    • Smathers, R.L.1    Galligan, J.J.2    Stewart, B.J.3    Petersen, D.R.4
  • 167
    • 40049104518 scopus 로고    scopus 로고
    • 4-Hydroxynonenal-protein adducts: A reliable biomarker of lipid oxidation in liver diseases
    • 2-s2.0-40049104518 10.1016/j.mam.2007.09.016
    • Poli G., Biasi F., Leonarduzzi G., 4-Hydroxynonenal-protein adducts: a reliable biomarker of lipid oxidation in liver diseases. Molecular Aspects of Medicine 2008 29 1-2 67 71 2-s2.0-40049104518 10.1016/j.mam.2007.09.016
    • (2008) Molecular Aspects of Medicine , vol.29 , Issue.1-2 , pp. 67-71
    • Poli, G.1    Biasi, F.2    Leonarduzzi, G.3
  • 168
    • 4444314070 scopus 로고    scopus 로고
    • Reactions of 4-hydroxynonenal with proteins and cellular targets
    • DOI 10.1016/j.freeradbiomed.2004.06.012, PII S0891584904004666
    • Petersen D. R., Doorn J. A., Reactions of 4-hydroxynonenal with proteins and cellular targets. Free Radical Biology and Medicine 2004 37 7 937 945 2-s2.0-4444314070 10.1016/j.freeradbiomed.2004.06.012 (Pubitemid 39164576)
    • (2004) Free Radical Biology and Medicine , vol.37 , Issue.7 , pp. 937-945
    • Petersen, D.R.1    Doorn, J.A.2
  • 170
    • 84866848255 scopus 로고    scopus 로고
    • 4-Hydroxy-2-nonenal, a reactive product of lipid peroxidation, and neurodegenerative diseases: A toxic combination illuminated by redox proteomics studies
    • Perluigi M., Coccia R., Butterfield D. A., 4-Hydroxy-2-nonenal, a reactive product of lipid peroxidation, and neurodegenerative diseases: a toxic combination illuminated by redox proteomics studies. Antioxidants & Redox Signaling 2012 17 11 1590 1609
    • (2012) Antioxidants & Redox Signaling , vol.17 , Issue.11 , pp. 1590-1609
    • Perluigi, M.1    Coccia, R.2    Butterfield, D.A.3
  • 171
    • 80052261937 scopus 로고    scopus 로고
    • Lipid peroxidation and neurodegenerative disease
    • 2-s2.0-80052261937 10.1016/j.freeradbiomed.2011.06.027
    • Reed T. T., Lipid peroxidation and neurodegenerative disease. Free Radical Biology and Medicine 2011 51 7 1302 1319 2-s2.0-80052261937 10.1016/j.freeradbiomed.2011.06.027
    • (2011) Free Radical Biology and Medicine , vol.51 , Issue.7 , pp. 1302-1319
    • Reed, T.T.1
  • 172
    • 0043172468 scopus 로고    scopus 로고
    • 4-Hydroxynonenal and neurodegenerative diseases
    • DOI 10.1016/S0098-2997(03)00024-4
    • Zarkovic K., 4-hydroxynonenal and neurodegenerative diseases. Molecular Aspects of Medicine 2003 24 4-5 293 303 2-s2.0-0043172468 10.1016/S0098-2997(03) 00024-4 (Pubitemid 36945029)
    • (2003) Molecular Aspects of Medicine , vol.24 , Issue.4-5 , pp. 293-303
    • Zarkovic, K.1
  • 173
    • 0032527641 scopus 로고    scopus 로고
    • (E)-4-Hydroxy-2-nonenal may be involved in the pathogenesis of Parkinson's disease
    • DOI 10.1016/S0891-5849(98)00021-5, PII S0891584998000215
    • Selley M. L., (E)-4-hydroxy-2-nonenal may be involved in the pathogenesis of Parkinson's disease. Free Radical Biology and Medicine 1998 25 2 169 174 2-s2.0-0032527641 10.1016/S0891-5849(98)00021-5 (Pubitemid 28293150)
    • (1998) Free Radical Biology and Medicine , vol.25 , Issue.2 , pp. 169-174
    • Selley, M.L.1
  • 176
    • 0034938943 scopus 로고    scopus 로고
    • Elucidation of reaction scheme describing malondialdehyde - Acetaldehyde - Protein adduct formation
    • DOI 10.1021/tx000222a
    • Tuma D. J., Kearley M. L., Thiele G. M., Worrall S., Haver A., Klassen L. W., Sorrell M. F., Elucidation of reaction scheme describing malondialdehyde-acetaldehyde-protein adduct formation. Chemical Research in Toxicology 2001 14 7 822 832 2-s2.0-0034938943 10.1021/tx000222a (Pubitemid 32661337)
    • (2001) Chemical Research in Toxicology , vol.14 , Issue.7 , pp. 822-832
    • Tuma, D.J.1    Kearley, M.L.2    Thiele, G.M.3    Worrall, S.4    Haver, A.5    Klassen, L.W.6    Sorrell, M.F.7
  • 177
    • 84868314089 scopus 로고    scopus 로고
    • Differential oxidative modification of proteins in MRL+/+ and MRL/lpr mice: Increased formation of lipid peroxidation-derived aldehyde-protein adducts may contribute to accelerated onset of autoimmune response
    • Wang G., Li H., Firoze Khan M., Differential oxidative modification of proteins in MRL+/+ and MRL/lpr mice: increased formation of lipid peroxidation-derived aldehyde-protein adducts may contribute to accelerated onset of autoimmune response. Free Radical Research 2012 46 12 1472 1481
    • (2012) Free Radical Research , vol.46 , Issue.12 , pp. 1472-1481
    • Wang, G.1    Li, H.2    Firoze Khan, M.3
  • 178
    • 50949129855 scopus 로고    scopus 로고
    • Increased immunogenicity to P815 cells modified with malondialdehyde and acetaldehyde
    • 2-s2.0-50949129855 10.1016/j.intimp.2008.03.022
    • Duryee M. J., Klassen L. W., Jones B. L., Willis M. S., Tuma D. J., Thiele G. M., Increased immunogenicity to P815 cells modified with malondialdehyde and acetaldehyde. International Immunopharmacology 2008 8 8 1112 1118 2-s2.0-50949129855 10.1016/j.intimp.2008.03.022
    • (2008) International Immunopharmacology , vol.8 , Issue.8 , pp. 1112-1118
    • Duryee, M.J.1    Klassen, L.W.2    Jones, B.L.3    Willis, M.S.4    Tuma, D.J.5    Thiele, G.M.6
  • 179
    • 35648977125 scopus 로고    scopus 로고
    • Involvement of lipid peroxidation-derived aldehyde-protein adducts in autoimmunity mediated by trichloroethene
    • DOI 10.1080/15287390701550888, PII 783547414
    • Wang G., Ansari G. A. S., Khan M. F., Involvement of lipid peroxidation-derived aldehyde-protein adducts in autoimmunity mediated by trichloroethene. Journal of Toxicology and Environmental Health A: Current Issues 2007 70 23 1977 1985 2-s2.0-35648977125 10.1080/15287390701550888 (Pubitemid 350034921)
    • (2007) Journal of Toxicology and Environmental Health - Part A: Current Issues , vol.70 , Issue.23 , pp. 1977-1985
    • Wang, G.1    Ansari, G.A.S.2    Khan, M.F.3
  • 180
    • 34447537998 scopus 로고    scopus 로고
    • Malondialdehyde modification of myelin oligodendrocyte glycoprotein leads to increased immunogenicity and encephalitogenicity
    • DOI 10.1002/eji.200636912
    • Wållberg M., Bergquist J., Achour A., Breij E., Harris R. A., Malondialdehyde modification of myelin oligodendrocyte glycoprotein leads to increased immunogenicity and encephalitogenicity. European Journal of Immunology 2007 37 7 1986 1995 2-s2.0-34447537998 10.1002/eji.200636912 (Pubitemid 47065134)
    • (2007) European Journal of Immunology , vol.37 , Issue.7 , pp. 1986-1995
    • Wallberg, M.1    Bergquist, J.2    Achour, A.3    Breij, E.4    Harris, R.A.5
  • 181
    • 84863986254 scopus 로고    scopus 로고
    • The innate immune response to products of phospholipid peroxidation
    • Weismann D., Binder C. J., The innate immune response to products of phospholipid peroxidation. Biochimica et Biophysica Acta: Biomembranes 2012 1818 10 2465 2475
    • (2012) Biochimica et Biophysica Acta: Biomembranes , vol.1818 , Issue.10 , pp. 2465-2475
    • Weismann, D.1    Binder, C.J.2
  • 182
    • 0347683483 scopus 로고    scopus 로고
    • Identification of a New Cross-link and Unique Histidine Adduct from Bovine Serum Albumin Incubated with Malondialdehyde
    • DOI 10.1074/jbc.M310608200
    • Slatter D. A., Avery N. C., Bailey A. J., Identification of a new cross-link and unique histidine adduct from bovine serum albumin incubated with malondialdehyde. Journal of Biological Chemistry 2004 279 1 61 69 2-s2.0-0347683483 10.1074/jbc.M310608200 (Pubitemid 38044799)
    • (2004) Journal of Biological Chemistry , vol.279 , Issue.1 , pp. 61-69
    • Slatter, D.A.1    Avery, N.C.2    Bailey, A.J.3
  • 183
    • 78649752112 scopus 로고    scopus 로고
    • The cytotoxic mechanism of malondialdehyde and protective effect of carnosine via protein cross-linking/mitochondrial dysfunction/reactive oxygen species/MAPK pathway in neurons
    • 2-s2.0-78649752112 10.1016/j.ejphar.2010.09.033
    • Cheng J., Wang F., Yu D.-F., Wu P.-F., Chen J.-G., The cytotoxic mechanism of malondialdehyde and protective effect of carnosine via protein cross-linking/mitochondrial dysfunction/reactive oxygen species/MAPK pathway in neurons. European Journal of Pharmacology 2011 650 1 184 194 2-s2.0-78649752112 10.1016/j.ejphar.2010.09.033
    • (2011) European Journal of Pharmacology , vol.650 , Issue.1 , pp. 184-194
    • Cheng, J.1    Wang, F.2    Yu, D.-F.3    Wu, P.-F.4    Chen, J.-G.5
  • 185
    • 79960958491 scopus 로고    scopus 로고
    • Specific recognition of malondialdehyde and malondialdehyde acetaldehyde adducts on oxidized LDL and apoptotic cells by complement anaphylatoxin C3a
    • 2-s2.0-79960958491 10.1016/j.freeradbiomed.2011.05.029
    • Veneskoski M., Turunen S. P., Kummu O., Nissinen A., Rannikko S., Levonen A.-L., Hörkkö S., Specific recognition of malondialdehyde and malondialdehyde acetaldehyde adducts on oxidized LDL and apoptotic cells by complement anaphylatoxin C3a. Free Radical Biology and Medicine 2011 51 4 834 843 2-s2.0-79960958491 10.1016/j.freeradbiomed.2011.05.029
    • (2011) Free Radical Biology and Medicine , vol.51 , Issue.4 , pp. 834-843
    • Veneskoski, M.1    Turunen, S.P.2    Kummu, O.3    Nissinen, A.4    Rannikko, S.5    Levonen, A.-L.6    Hörkkö, S.7
  • 186
    • 0036471578 scopus 로고    scopus 로고
    • Effect of malondialdehyde-acetaldehyde-protein adducts on the protein kinase C-dependent secretion of urokinase-type plasminogen activator in hepatic stellate cells
    • DOI 10.1016/S0006-2952(01)00883-8, PII S0006295201008838
    • Kharbanda K. K., Shubert K. A., Wyatt T. A., Sorrell M. F., Tuma D. J., Effect of malondialdehyde-acetaldehyde-protein adducts on the protein kinase C-dependent secretion of urokinase-type plasminogen activator in hepatic stellate cells. Biochemical Pharmacology 2002 63 3 553 562 2-s2.0-0036471578 10.1016/S0006-2952(01)00883-8 (Pubitemid 34169735)
    • (2002) Biochemical Pharmacology , vol.63 , Issue.3 , pp. 553-562
    • Kharbanda, K.K.1    Shubert, K.A.2    Wyatt, T.A.3    Sorrell, M.F.4    Tuma, D.J.5
  • 187
    • 18144452679 scopus 로고    scopus 로고
    • Oxy radicals, lipid peroxidation and DNA damage
    • DOI 10.1016/S0300-483X(02)00448-1, PII S0300483X02004481
    • Marnett L. J., Oxy radicals, lipid peroxidation and DNA damage. Toxicology 2002 181-182 219 222 2-s2.0-18144452679 10.1016/S0300-483X(02)00448-1 (Pubitemid 36015702)
    • (2002) Toxicology , vol.181-182 , pp. 219-222
    • Marnett, L.J.1
  • 188
    • 0033535371 scopus 로고    scopus 로고
    • Lipid peroxidation-DNA damage by malondialdehyde
    • Marnett L. J., Lipid peroxidation-DNA damage by malondialdehyde. Mutation Research 1999 424 1-2 83 95
    • (1999) Mutation Research , vol.424 , Issue.1-2 , pp. 83-95
    • Marnett, L.J.1
  • 189
    • 0030744202 scopus 로고    scopus 로고
    • Mutagenicity in Escherichia coli of the major DNA adduct derived from the endogenous mutagen malondialdehyde
    • DOI 10.1073/pnas.94.16.8652
    • Fink S. P., Reddy G. R., Marnett L. J., Mutagenicity in Escherichia coli of the major DNA adduct derived from the endogenous mutagen malondialdehyde. Proceedings of the National Academy of Sciences of the United States of America 1997 94 16 8652 8657 2-s2.0-0030744202 10.1073/pnas.94.16.8652 (Pubitemid 27335094)
    • (1997) Proceedings of the National Academy of Sciences of the United States of America , vol.94 , Issue.16 , pp. 8652-8657
    • Fink, S.P.1    Reddy, G.R.2    Marnett, L.J.3
  • 190
    • 0344589347 scopus 로고    scopus 로고
    • Synergistic DNA damaging effects of malondialdehyde/Cu(II) in PM2 DNA and in human fibroblasts
    • DOI 10.1016/S0378-4274(98)00002-2, PII S0378427498000022
    • Vöhringer M.-L., Becker T. W., Krieger G., Jacobi H., Witte I., Synergistic DNA damaging effects of malondialdehyde/Cu(II) in PM2 DNA and in human fibroblasts. Toxicology Letters 1998 94 3 159 166 2-s2.0-0344589347 10.1016/S0378-4274(98)00002-2 (Pubitemid 28230995)
    • (1998) Toxicology Letters , vol.94 , Issue.3 , pp. 159-166
    • Vohringer, M.-L.1    Becker, T.W.2    Krieger, G.3    Jacobi, H.4    Witte, I.5
  • 191
    • 0031878858 scopus 로고    scopus 로고
    • Induction of cell cycle arrest by the endogenous product of lipid peroxidation, malondialdehyde
    • DOI 10.1093/carcin/19.7.1275
    • Ji C., Rouzer C. A., Marnett L. J., Pietenpol J. A., Induction of cell cycle arrest by the endogenous product of lipid peroxidation, malondialdehyde. Carcinogenesis 1998 19 7 1275 1283 2-s2.0-0031878858 10.1093/carcin/19.7.1275 (Pubitemid 28375380)
    • (1998) Carcinogenesis , vol.19 , Issue.7 , pp. 1275-1283
    • Ji, C.1    Rouzer, C.A.2    Marnett, L.J.3    Pietenpol, J.A.4
  • 192
    • 2942522656 scopus 로고    scopus 로고
    • Malondialdehyde-acetaldehyde haptenated protein binds macrophage scavenger receptor(s) and induces lysosomal damage
    • DOI 10.1016/j.intimp.2004.04.004, PII S1567576904001171
    • Willis M. S., Klassen L. W., Carlson D. L., Brouse C. F., Thiele G. M., Malondialdehyde-acetaldehyde haptenated protein binds macrophage scavenger receptor(s) and induces lysosomal damage. International Immunopharmacology 2004 4 7 885 899 2-s2.0-2942522656 10.1016/j.intimp.2004.04.004 (Pubitemid 38739094)
    • (2004) International Immunopharmacology , vol.4 , Issue.7 , pp. 885-899
    • Willis, M.S.1    Klassen, L.W.2    Carlson, D.L.3    Brouse, C.F.4    Thiele, G.M.5
  • 194
    • 78649839856 scopus 로고    scopus 로고
    • Arrest of human mitochondrial RNA polymerase transcription by the biological aldehyde adduct of DNA, M1dG
    • 2-s2.0-78649839856 10.1093/nar/gkq656
    • Cline S. D., Lodeiro M. F., Marnett L. J., Cameron C. E., Arnold J. J., Arrest of human mitochondrial RNA polymerase transcription by the biological aldehyde adduct of DNA, M1dG. Nucleic Acids Research 2010 38 21 7546 7557 2-s2.0-78649839856 10.1093/nar/gkq656
    • (2010) Nucleic Acids Research , vol.38 , Issue.21 , pp. 7546-7557
    • Cline, S.D.1    Lodeiro, M.F.2    Marnett, L.J.3    Cameron, C.E.4    Arnold, J.J.5
  • 196
    • 84878884100 scopus 로고    scopus 로고
    • The lipid peroxidation product 4-hydroxy-2-nonenal: Advances in chemistry and analysis
    • Spickett C. M., The lipid peroxidation product 4-hydroxy-2-nonenal: advances in chemistry and analysis. Redox Biology 2013 1 1 145 152
    • (2013) Redox Biology , vol.1 , Issue.1 , pp. 145-152
    • Spickett, C.M.1
  • 197
    • 82955237301 scopus 로고    scopus 로고
    • Hydroxyalkenals and oxidized phospholipids modulation of endothelial cytoskeleton, focal adhesion and adherens junction proteins in regulating endothelial barrier function
    • 2-s2.0-82955237301 10.1016/j.mvr.2011.04.012
    • Usatyuk P. V., Natarajan V., Hydroxyalkenals and oxidized phospholipids modulation of endothelial cytoskeleton, focal adhesion and adherens junction proteins in regulating endothelial barrier function. Microvascular Research 2012 83 1 45 55 2-s2.0-82955237301 10.1016/j.mvr.2011.04.012
    • (2012) Microvascular Research , vol.83 , Issue.1 , pp. 45-55
    • Usatyuk, P.V.1    Natarajan, V.2
  • 199
    • 80051801444 scopus 로고    scopus 로고
    • Relationship of electrophilic stress to aging
    • 2-s2.0-80051801444 10.1016/j.freeradbiomed.2011.05.039
    • Zimniak P., Relationship of electrophilic stress to aging. Free Radical Biology and Medicine 2011 51 6 1087 1105 2-s2.0-80051801444 10.1016/j. freeradbiomed.2011.05.039
    • (2011) Free Radical Biology and Medicine , vol.51 , Issue.6 , pp. 1087-1105
    • Zimniak, P.1
  • 200
    • 80052998418 scopus 로고    scopus 로고
    • Exploring the biology of lipid peroxidation-derived protein carbonylation
    • Fritz K. S., Petersen D. R., Exploring the biology of lipid peroxidation-derived protein carbonylation. Chemical Research in Toxicology 2011 24 9 1411 1419
    • (2011) Chemical Research in Toxicology , vol.24 , Issue.9 , pp. 1411-1419
    • Fritz, K.S.1    Petersen, D.R.2
  • 201
    • 78650147789 scopus 로고    scopus 로고
    • Roles of 3-nitrotyrosine- and 4-hydroxynonenal-modified brain proteins in the progression and pathogenesis of Alzheimer's disease
    • 2-s2.0-78650147789 10.3109/10715762.2010.520014
    • Butterfield D. A., Reed T., Sultana R., Roles of 3-nitrotyrosine- and 4-hydroxynonenal-modified brain proteins in the progression and pathogenesis of Alzheimer's disease. Free Radical Research 2011 45 1 59 72 2-s2.0-78650147789 10.3109/10715762.2010.520014
    • (2011) Free Radical Research , vol.45 , Issue.1 , pp. 59-72
    • Butterfield, D.A.1    Reed, T.2    Sultana, R.3
  • 202
    • 79954621602 scopus 로고    scopus 로고
    • Interactions of glutathione transferases with 4-hydroxynonenal
    • Balogh L. M., Atkins W. M., Interactions of glutathione transferases with 4-hydroxynonenal. Drug Metabolism Reviews 2011 43 2 165 178
    • (2011) Drug Metabolism Reviews , vol.43 , Issue.2 , pp. 165-178
    • Balogh, L.M.1    Atkins, W.M.2
  • 206
    • 0030603977 scopus 로고    scopus 로고
    • Oxygenation of (3Z)-alkenal to (2E)-4-hydroxy-2-alkenal in soybean seed (Glycine max L.)
    • DOI 10.1016/0005-2760(96)00076-8
    • Takamura H., Gardner H. W., Oxygenation of (3Z)-alkenal to (2E)-4-hydroxy-2-alkenal in soybean seed (Glycine max L.). Biochimica et Biophysica Acta: Lipids and Lipid Metabolism 1996 1303 2 83 91 2-s2.0-0030603977 10.1016/0005-2760(96)00076-8 (Pubitemid 26325538)
    • (1996) Biochimica et Biophysica Acta - Lipids and Lipid Metabolism , vol.1303 , Issue.2 , pp. 83-91
    • Takamura, H.1    Gardner, H.W.2
  • 207
    • 0035877699 scopus 로고    scopus 로고
    • Two distinct pathways of formation of 4-hydroxynonenal. Mechanisms of nonenzymatic transformation of the 9- and 13-hydroperoxides of linoleic acid to 4-hydroxyalkenals
    • 2-s2.0-0035877699 10.1074/jbc.M101821200
    • Schneider C., Tallman K. A., Porter N. A., Brash A. R., Two distinct pathways of formation of 4-hydroxynonenal. Mechanisms of nonenzymatic transformation of the 9- and 13-hydroperoxides of linoleic acid to 4-hydroxyalkenals. Journal of Biological Chemistry 2001 276 24 20831 20838 2-s2.0-0035877699 10.1074/jbc.M101821200
    • (2001) Journal of Biological Chemistry , vol.276 , Issue.24 , pp. 20831-20838
    • Schneider, C.1    Tallman, K.A.2    Porter, N.A.3    Brash, A.R.4
  • 209
    • 34249980917 scopus 로고    scopus 로고
    • Effect of 15-lipoxygenase metabolites, 15-(S)-HPETE and 15-(S)-HETE on chronic myelogenous leukemia cell line K-562: Reactive oxygen species (ROS) mediate caspase-dependent apoptosis
    • DOI 10.1016/j.bcp.2007.04.005, PII S0006295207002237
    • Mahipal S. V. K., Subhashini J., Reddy M. C., Reddy M. M., Anilkumar K., Roy K. R., Reddy G. V., Reddanna P., Effect of 15-lipoxygenase metabolites, 15-(S)-HPETE and 15-(S)-HETE on chronic myelogenous leukemia cell line K-562: reactive oxygen species (ROS) mediate caspase-dependent apoptosis. Biochemical Pharmacology 2007 74 2 202 214 2-s2.0-34249980917 10.1016/j.bcp.2007.04.005 (Pubitemid 46890813)
    • (2007) Biochemical Pharmacology , vol.74 , Issue.2 , pp. 202-214
    • Mahipal, S.V.K.1    Subhashini, J.2    Reddy, M.C.3    Reddy, M.M.4    Anilkumar, K.5    Roy, K.R.6    Reddy, G.V.7    Reddanna, P.8
  • 210
    • 60549093622 scopus 로고    scopus 로고
    • Effects of (15S)-hydroperoxyeicosatetraenoic acid and (15S)-hydroxyeicosatetraenoic acid on the acute-lymphoblastic-leukaemia cell line Jurkat: Activation of the Fas-mediated death pathway
    • 2-s2.0-60549093622 10.1042/BA20070264
    • Kumar K. A., Arunasree K. M., Roy K. R., Reddy N. P., Aparna A., Reddy G. V., Reddanna P., Effects of (15S)-hydroperoxyeicosatetraenoic acid and (15S)-hydroxyeicosatetraenoic acid on the acute-lymphoblastic-leukaemia cell line Jurkat: activation of the Fas-mediated death pathway. Biotechnology and Applied Biochemistry 2009 52 2 121 133 2-s2.0-60549093622 10.1042/BA20070264
    • (2009) Biotechnology and Applied Biochemistry , vol.52 , Issue.2 , pp. 121-133
    • Kumar, K.A.1    Arunasree, K.M.2    Roy, K.R.3    Reddy, N.P.4    Aparna, A.5    Reddy, G.V.6    Reddanna, P.7
  • 211
    • 0042671405 scopus 로고    scopus 로고
    • Genotoxicity of HNE
    • DOI 10.1016/S0098-2997(03)00010-4
    • Eckl P. M., Genotoxicity of HNE. Molecular Aspects of Medicine 2003 24 4-5 161 165 (Pubitemid 36945016)
    • (2003) Molecular Aspects of Medicine , vol.24 , Issue.4-5 , pp. 161-165
    • Eckl, P.M.1
  • 212
    • 0042170296 scopus 로고    scopus 로고
    • Intracellular metabolism of 4-hydroxynonenal
    • DOI 10.1016/S0098-2997(03)00011-6
    • Siems W., Grune T., Intracellular metabolism of 4-hydroxynonenal. Molecular Aspects of Medicine 2003 24 4-5 167 175 (Pubitemid 36945017)
    • (2003) Molecular Aspects of Medicine , vol.24 , Issue.4-5 , pp. 167-175
    • Siems, W.1    Grune, T.2
  • 213
    • 0041669529 scopus 로고    scopus 로고
    • Fate of 4-hydroxynonenal in vivo: Disposition and metabolic pathways
    • DOI 10.1016/S0098-2997(03)00012-8
    • Alary J., Guéraud F., Cravedi J.-P., Fate of 4-hydroxynonenal in vivo: disposition and metabolic pathways. Molecular Aspects of Medicine 2003 24 4-5 177 187 2-s2.0-0041669529 10.1016/S0098-2997(03)00012-8 (Pubitemid 36945018)
    • (2003) Molecular Aspects of Medicine , vol.24 , Issue.4-5 , pp. 177-187
    • Alary, J.1    Gueraud, F.2    Cravedi, J.-P.3
  • 214
    • 84901934722 scopus 로고    scopus 로고
    • Gsta4 null mouse embryonic fibroblasts exhibit enhanced sensitivity to oxidants: Role of 4-hydroxynonenal in oxidant toxicity
    • McElhanon K. E., Bose C., Sharma R., Wu L., Awasthi Y. C., Singh S. P., Gsta4 null mouse embryonic fibroblasts exhibit enhanced sensitivity to oxidants: role of 4-hydroxynonenal in oxidant toxicity. Open Journal of Apoptosis 2013 2 1
    • (2013) Open Journal of Apoptosis , vol.2 , Issue.1
    • McElhanon, K.E.1    Bose, C.2    Sharma, R.3    Wu, L.4    Awasthi, Y.C.5    Singh, S.P.6
  • 215
    • 84859505181 scopus 로고    scopus 로고
    • Molecular mechanisms of ALDH3A1-mediated cellular protection against 4-hydroxy-2-nonenal
    • 2-s2.0-84859505181 10.1016/j.freeradbiomed.2012.02.050
    • Black W., Chen Y., Matsumoto A., Thompson D. C., Lassen N., Pappa A., Vasiliou V., Molecular mechanisms of ALDH3A1-mediated cellular protection against 4-hydroxy-2-nonenal. Free Radical Biology and Medicine 2012 52 9 1937 1944 2-s2.0-84859505181 10.1016/j.freeradbiomed.2012.02.050
    • (2012) Free Radical Biology and Medicine , vol.52 , Issue.9 , pp. 1937-1944
    • Black, W.1    Chen, Y.2    Matsumoto, A.3    Thompson, D.C.4    Lassen, N.5    Pappa, A.6    Vasiliou, V.7
  • 216
    • 84863867764 scopus 로고    scopus 로고
    • Modulation of aldehyde dehydrogenase activity affects (±)-4-hydroxy-2E-nonenal (HNE) toxicity and HNE-protein adduct levels in PC12 cells
    • 2-s2.0-83255189605 10.1007/s12031-011-9688-y
    • Kong D., Kotraiah V., Modulation of aldehyde dehydrogenase activity affects (±)-4-hydroxy-2E-nonenal (HNE) toxicity and HNE-protein adduct levels in PC12 cells. Journal of Molecular Neuroscience 2012 47 3 595 603 2-s2.0-83255189605 10.1007/s12031-011-9688-y
    • (2012) Journal of Molecular Neuroscience , vol.47 , Issue.3 , pp. 595-603
    • Kong, D.1    Kotraiah, V.2
  • 217
    • 84870920138 scopus 로고    scopus 로고
    • Anti-oxidative stress regulator NF-E2-related factor 2 mediates the adaptive induction of antioxidant and detoxifying enzymes by lipid peroxidation metabolite 4-hydroxynonenal
    • ARTICLE 40
    • Huang Y., Li W., Kong A. N. T., Anti-oxidative stress regulator NF-E2-related factor 2 mediates the adaptive induction of antioxidant and detoxifying enzymes by lipid peroxidation metabolite 4-hydroxynonenal. Cell & Bioscience 2012 2 1, article 40
    • (2012) Cell & Bioscience , vol.2 , Issue.1
    • Huang, Y.1    Li, W.2    Kong, A.N.T.3
  • 219
    • 33846809497 scopus 로고    scopus 로고
    • Modulation of antioxidant gene expression by 4-hydroxynonenal: Atheroprotective role of the Nrf2/ARE transcription pathway
    • DOI 10.1179/135100007X162167
    • Siow R. C. M., Ishii T., Mann G. E., Modulation of antioxidant gene expression by 4-hydroxynonenal: atheroprotective role of the Nrf2/ARE transcription pathway. Redox Report 2007 12 1-2 11 15 2-s2.0-33846809497 10.1179/135100007X162167 (Pubitemid 46210347)
    • (2007) Redox Report , vol.12 , Issue.1-2 , pp. 11-15
    • Siow, R.C.M.1    Ishii, T.2    Mann, G.E.3
  • 220
    • 34248591499 scopus 로고    scopus 로고
    • Upregulation of thioredoxin system via Nrf2-antioxidant responsive element pathway in adaptive-retinal neuroprotection in vivo and in vitro
    • DOI 10.1016/j.freeradbiomed.2007.03.018, PII S0891584907002146
    • Tanito M., Agbaga M.-P., Anderson R. E., Upregulation of thioredoxin system via Nrf2-antioxidant responsive element pathway in adaptive-retinal neuroprotection in vivo and in vitro. Free Radical Biology and Medicine 2007 42 12 1838 1850 2-s2.0-34248591499 10.1016/j.freeradbiomed.2007.03.018 (Pubitemid 46754760)
    • (2007) Free Radical Biology and Medicine , vol.42 , Issue.12 , pp. 1838-1850
    • Tanito, M.1    Agbaga, M.-P.2    Anderson, R.E.3
  • 221
    • 1642396537 scopus 로고    scopus 로고
    • Role of Nrf2 in the Regulation of CD36 and Stress Protein Expression in Murine Macrophages: Activation by Oxidatively Modified LDL and 4-Hydroxynonenal
    • DOI 10.1161/01.RES.0000119171.44657.45
    • Ishii T., Itoh K., Ruiz E., Leake D. S., Unoki H., Yamamoto M., Mann G. E., Role of Nrf2 in the regulation of CD36 and stress protein expression in murine macrophages: activation by oxidatively modified LDL and 4-hydroxynonenal. Circulation Research 2004 94 5 609 616 2-s2.0-1642396537 10.1161/01.RES. 0000119171.44657.45 (Pubitemid 38387743)
    • (2004) Circulation Research , vol.94 , Issue.5 , pp. 609-616
    • Ishii, T.1    Itoh, K.2    Ruiz, E.3    Leake, D.S.4    Unoki, H.5    Yamamoto, M.6    Mann, G.E.7
  • 222
    • 84874633349 scopus 로고    scopus 로고
    • Administration of the Nrf2-ARE activators sulforaphane and carnosic acid attenuates 4-hydroxy-2-nonenal-induced mitochondrial dysfunction ex vivo
    • Miller D. M., Singh I. N., Wang J. A., Hall E. D., Administration of the Nrf2-ARE activators sulforaphane and carnosic acid attenuates 4-hydroxy-2-nonenal-induced mitochondrial dysfunction ex vivo. Free Radical Biology and Medicine 2013 57 1 9
    • (2013) Free Radical Biology and Medicine , vol.57 , pp. 1-9
    • Miller, D.M.1    Singh, I.N.2    Wang, J.A.3    Hall, E.D.4
  • 223
  • 224
    • 84890940231 scopus 로고    scopus 로고
    • Oncogenic potential of Nrf2 and its principal target protein heme oxygenase-1
    • Na H. K., Surh Y. J., Oncogenic potential of Nrf2 and its principal target protein heme oxygenase-1. Free Radical Biology and Medicine 2014 67 353 365
    • (2014) Free Radical Biology and Medicine , vol.67 , pp. 353-365
    • Na, H.K.1    Surh, Y.J.2
  • 225
    • 84884867438 scopus 로고    scopus 로고
    • The role of Nrf2: Adipocyte differentiation, obesity, and insulin resistance
    • 184598 10.1155/2013/184598
    • Seo H. A., Lee I. K., The role of Nrf2: adipocyte differentiation, obesity, and insulin resistance. Oxidative Medicine and Cellular Longevity 2013 2013 7 184598 10.1155/2013/184598
    • (2013) Oxidative Medicine and Cellular Longevity , vol.2013 , pp. 7
    • Seo, H.A.1    Lee, I.K.2
  • 226
    • 84876788364 scopus 로고    scopus 로고
    • Regulation of oxidative stress by Nrf2 in the pathophysiology of infectious diseases
    • Deramaudt T. B., Dill C., Bonay M., Regulation of oxidative stress by Nrf2 in the pathophysiology of infectious diseases. Médecine et Maladies Infectieuses 2013 43 3 100 107
    • (2013) Médecine et Maladies Infectieuses , vol.43 , Issue.3 , pp. 100-107
    • Deramaudt, T.B.1    Dill, C.2    Bonay, M.3
  • 227
    • 84856380036 scopus 로고    scopus 로고
    • Haem oxygenase-1: Non-canonical roles in physiology and pathology
    • 2-s2.0-84856380036 10.1042/CS20110147
    • Grochot-Przeczek A., Dulak J., Jozkowicz A., Haem oxygenase-1: non-canonical roles in physiology and pathology. Clinical Science 2012 122 3 93 103 2-s2.0-84856380036 10.1042/CS20110147
    • (2012) Clinical Science , vol.122 , Issue.3 , pp. 93-103
    • Grochot-Przeczek, A.1    Dulak, J.2    Jozkowicz, A.3
  • 228
    • 84890138793 scopus 로고    scopus 로고
    • Lipid peroxidation end product 4-hydroxy-trans-2-nonenal triggers unfolded protein response and heme oxygenase-1 expression in PC12 cells: Roles of ROS and MAPK pathways
    • Lin M. H., Yen J. H., Weng C. Y., Wang L., Ha C. L., Wu M. J., Lipid peroxidation end product 4-hydroxy-trans-2-nonenal triggers unfolded protein response and heme oxygenase-1 expression in PC12 cells: roles of ROS and MAPK pathways. Toxicology 2014 315 24 37
    • (2014) Toxicology , vol.315 , pp. 24-37
    • Lin, M.H.1    Yen, J.H.2    Weng, C.Y.3    Wang, L.4    Ha, C.L.5    Wu, M.J.6
  • 229
    • 78049269927 scopus 로고    scopus 로고
    • Low concentration of 4-hydroxy hexenal increases heme oxygenase-1 expression through activation of Nrf2 and antioxidative activity in vascular endothelial cells
    • 2-s2.0-78049269927 10.1016/j.bbrc.2010.09.124
    • Ishikado A., Nishio Y., Morino K., Ugi S., Kondo H., Makino T., Kashiwagi A., Maegawa H., Low concentration of 4-hydroxy hexenal increases heme oxygenase-1 expression through activation of Nrf2 and antioxidative activity in vascular endothelial cells. Biochemical and Biophysical Research Communications 2010 402 1 99 104 2-s2.0-78049269927 10.1016/j.bbrc.2010.09.124
    • (2010) Biochemical and Biophysical Research Communications , vol.402 , Issue.1 , pp. 99-104
    • Ishikado, A.1    Nishio, Y.2    Morino, K.3    Ugi, S.4    Kondo, H.5    Makino, T.6    Kashiwagi, A.7    Maegawa, H.8
  • 231
    • 77952311188 scopus 로고    scopus 로고
    • Thioredoxin and thioredoxin reductase: Current research with special reference to human disease
    • 2-s2.0-77952311188 10.1016/j.bbrc.2010.03.083
    • Holmgren A., Lu J., Thioredoxin and thioredoxin reductase: current research with special reference to human disease. Biochemical and Biophysical Research Communications 2010 396 1 120 124 2-s2.0-77952311188 10.1016/j.bbrc.2010.03.083
    • (2010) Biochemical and Biophysical Research Communications , vol.396 , Issue.1 , pp. 120-124
    • Holmgren, A.1    Lu, J.2
  • 232
    • 29644437160 scopus 로고    scopus 로고
    • 4-Hydroxynonenal induces adaptive response and enhances PC12 cell tolerance primarily through induction of thioredoxin reductase 1 via activation of Nrf2
    • DOI 10.1074/jbc.M508556200
    • Chen Z.-H., Saito Y., Yoshida Y., Sekine A., Noguchi N., Niki E., 4-hydroxynonenal induces adaptive response and enhances PC12 cell tolerance primarily through induction of thioredoxin reductase 1 via activation of Nrf2. Journal of Biological Chemistry 2005 280 51 41921 41927 2-s2.0-29644437160 10.1074/jbc.M508556200 (Pubitemid 43023161)
    • (2005) Journal of Biological Chemistry , vol.280 , Issue.51 , pp. 41921-41927
    • Chen, Z.-H.1    Saito, Y.2    Yoshida, Y.3    Sekine, A.4    Noguchi, N.5    Niki, E.6
  • 233
    • 84875744148 scopus 로고    scopus 로고
    • Glutathione synthesis
    • Lu S. C., Glutathione synthesis. Biochimica et Biophysica Acta 2013 1830 5 3143 3153
    • (2013) Biochimica et Biophysica Acta , vol.1830 , Issue.5 , pp. 3143-3153
    • Lu, S.C.1
  • 234
    • 65049090510 scopus 로고    scopus 로고
    • Structure, function, and post-translational regulation of the catalytic and modifier subunits of glutamate cysteine ligase
    • 2-s2.0-65049090510 10.1016/j.mam.2008.08.009
    • Franklin C. C., Backos D. S., Mohar I., White C. C., Forman H. J., Kavanagh T. J., Structure, function, and post-translational regulation of the catalytic and modifier subunits of glutamate cysteine ligase. Molecular Aspects of Medicine 2009 30 1-2 86 98 2-s2.0-65049090510 10.1016/j.mam.2008.08.009
    • (2009) Molecular Aspects of Medicine , vol.30 , Issue.1-2 , pp. 86-98
    • Franklin, C.C.1    Backos, D.S.2    Mohar, I.3    White, C.C.4    Forman, H.J.5    Kavanagh, T.J.6
  • 235
    • 78650692177 scopus 로고    scopus 로고
    • Posttranslational modification and regulation of glutamate-cysteine ligase by the α, β -unsaturated aldehyde 4-hydroxy-2-nonenal
    • 2-s2.0-78650692177 10.1016/j.freeradbiomed.2010.10.694
    • Backos D. S., Fritz K. S., Roede J. R., Petersen D. R., Franklin C. C., Posttranslational modification and regulation of glutamate-cysteine ligase by the α β -unsaturated aldehyde 4-hydroxy-2-nonenal. Free Radical Biology and Medicine 2011 50 1 14 26 2-s2.0-78650692177 10.1016/j.freeradbiomed. 2010.10.694
    • (2011) Free Radical Biology and Medicine , vol.50 , Issue.1 , pp. 14-26
    • Backos, D.S.1    Fritz, K.S.2    Roede, J.R.3    Petersen, D.R.4    Franklin, C.C.5
  • 236
    • 57449083031 scopus 로고    scopus 로고
    • Resveratrol and 4-hydroxynonenal act in concert to increase glutamate cysteine ligase expression and glutathione in human bronchial epithelial cells
    • 2-s2.0-57449083031 10.1016/j.abb.2008.10.020
    • Zhang H., Shih A., Rinna A., Forman H. J., Resveratrol and 4-hydroxynonenal act in concert to increase glutamate cysteine ligase expression and glutathione in human bronchial epithelial cells. Archives of Biochemistry and Biophysics 2009 481 1 110 115 2-s2.0-57449083031 10.1016/j.abb.2008.10.020
    • (2009) Archives of Biochemistry and Biophysics , vol.481 , Issue.1 , pp. 110-115
    • Zhang, H.1    Shih, A.2    Rinna, A.3    Forman, H.J.4
  • 237
    • 33846849624 scopus 로고    scopus 로고
    • Submicromolar concentrations of 4-hydroxynonenal induce glutamate cysteine ligase expression in HBE1 cells
    • DOI 10.1179/135100007X162266
    • Zhang H., Court N., Forman H. J., Submicromolar concentrations of 4-hydroxynonenal induce glutamate cysteine ligase expression in HBE1 cells. Redox Report 2007 12 1-2 101 106 2-s2.0-33846849624 10.1179/135100007X162266 (Pubitemid 46210366)
    • (2007) Redox Report , vol.12 , Issue.1-2 , pp. 101-106
    • Zhang, H.1    Court, N.2    Forman, H.J.3
  • 238
    • 15744393896 scopus 로고    scopus 로고
    • Mechanisms of glutamate cysteine ligase (GCL) induction by 4-hydroxynonenal
    • DOI 10.1016/j.freeradbiomed.2004.11.012
    • Iles K. E., Liu R.-M., Mechanisms of Glutamate Cysteine Ligase (GCL) induction by 4-hydroxynonenal. Free Radical Biology and Medicine 2005 38 5 547 556 2-s2.0-15744393896 10.1016/j.freeradbiomed.2004.11.012 (Pubitemid 40416850)
    • (2005) Free Radical Biology and Medicine , vol.38 , Issue.5 , pp. 547-556
    • Iles, K.E.1    Liu, R.-M.2
  • 239
    • 0041625928 scopus 로고    scopus 로고
    • HNE - Signaling pathways leading to its elimination
    • DOI 10.1016/S0098-2997(03)00013-X
    • Forman H. J., Dickinson D. A., Iles K. E., HNE-signaling pathways leading to its elimination. Molecular Aspects of Medicine 2003 24 4-5 189 194 2-s2.0-0041625928 10.1016/S0098-2997(03)00013-X (Pubitemid 36957831)
    • (2003) Molecular Aspects of Medicine , vol.24 , Issue.4-5 , pp. 189-194
    • Forman, H.J.1    Dickinson, D.A.2    Iles, K.E.3
  • 240
    • 77958577388 scopus 로고    scopus 로고
    • Activation of metallothionein transcription by 4-hydroxynonenal
    • 2-s2.0-77958577388 10.1002/jbt.20342
    • Braithwaite E. K., Mattie M. D., Freedman J. H., Activation of metallothionein transcription by 4-hydroxynonenal. Journal of Biochemical and Molecular Toxicology 2010 24 5 330 334 2-s2.0-77958577388 10.1002/jbt.20342
    • (2010) Journal of Biochemical and Molecular Toxicology , vol.24 , Issue.5 , pp. 330-334
    • Braithwaite, E.K.1    Mattie, M.D.2    Freedman, J.H.3
  • 241
    • 4444233380 scopus 로고    scopus 로고
    • Hepatic stellate cells lack AP-1 responsiveness to electrophiles and phorbol 12-myristate-13-acetate
    • DOI 10.1016/j.bbrc.2004.07.180, PII S0006291X04017164
    • Reichard J. F., Petersen D. R., Hepatic stellate cells lack AP-1 responsiveness to electrophiles and phorbol 12-myristate-13-acetate. Biochemical and Biophysical Research Communications 2004 322 3 842 853 2-s2.0-4444233380 10.1016/j.bbrc.2004.07.180 (Pubitemid 39164429)
    • (2004) Biochemical and Biophysical Research Communications , vol.322 , Issue.3 , pp. 842-853
    • Reichard, J.F.1    Petersen, D.R.2
  • 242
    • 4344659607 scopus 로고    scopus 로고
    • 4-Hydroxy-2, 3-nonenal activates activator protein-1 and mitogen-activated protein kinases in rat pancreatic stellate cells
    • Kikuta K., Masamune A., Satoh M., Suzuki N., Shimosegawa T., 4-Hydroxy-2, 3-nonenal activates activator protein-1 and mitogen-activated protein kinases in rat pancreatic stellate cells. World Journal of Gastroenterology 2004 10 16 2344 2351 2-s2.0-4344659607 (Pubitemid 39157757)
    • (2004) World Journal of Gastroenterology , vol.10 , Issue.16 , pp. 2344-2351
    • Kikuta, K.1    Masamune, A.2    Satoh, M.3    Suzuki, N.4    Shimosegawa, T.5
  • 243
    • 0033985185 scopus 로고    scopus 로고
    • The lipid peroxidation product 4-hydroxy-2,3-nonenal increases AP-1- binding activity through caspase activation in neurons
    • DOI 10.1046/j.1471-4159.2000.0740159.x
    • Camandola S., Poli G., Mattson M. P., The lipid peroxidation product 4-hydroxy-2,3-nonenal increases AP-1- binding activity through caspase activation in neurons. Journal of Neurochemistry 2000 74 1 159 168 2-s2.0-0033985185 10.1046/j.1471-4159.2000.0740159.x (Pubitemid 30012766)
    • (2000) Journal of Neurochemistry , vol.74 , Issue.1 , pp. 159-168
    • Camandola, S.1    Poli, G.2    Mattson, M.P.3
  • 244
    • 0036098552 scopus 로고    scopus 로고
    • AP-1 as a regulator of cell life and death
    • DOI 10.1038/ncb0502-e131
    • Shaulian E., Karin M., AP-1 as a regulator of cell life and death. Nature Cell Biology 2002 4 5 E131 E136 2-s2.0-0036098552 10.1038/ncb0502-e131 (Pubitemid 34521026)
    • (2002) Nature Cell Biology , vol.4 , Issue.5
    • Shaulian, E.1    Karin, M.2
  • 245
    • 77949916262 scopus 로고    scopus 로고
    • AP-1 - The Jun proteins: Oncogenes or tumor suppressors in disguise?
    • 2-s2.0-77949916262 10.1016/j.cellsig.2009.12.008
    • Shaulian E., AP-1-the Jun proteins: oncogenes or tumor suppressors in disguise? Cellular Signalling 2010 22 6 894 899 2-s2.0-77949916262 10.1016/j.cellsig.2009.12.008
    • (2010) Cellular Signalling , vol.22 , Issue.6 , pp. 894-899
    • Shaulian, E.1
  • 246
    • 78650894319 scopus 로고    scopus 로고
    • Crosstalk of reactive oxygen species and NF- κ B signaling
    • Morgan M. J., Liu Z., Crosstalk of reactive oxygen species and NF- κ B signaling. Cell Research 2011 21 1 103 115
    • (2011) Cell Research , vol.21 , Issue.1 , pp. 103-115
    • Morgan, M.J.1    Liu, Z.2
  • 247
    • 84873732176 scopus 로고    scopus 로고
    • NF- κ B signaling pathway and free radical impact
    • Siomek A., NF- κ B signaling pathway and free radical impact. Acta Biochimica Polonica 2012 59 3 323 331
    • (2012) Acta Biochimica Polonica , vol.59 , Issue.3 , pp. 323-331
    • Siomek, A.1
  • 248
    • 33644946838 scopus 로고    scopus 로고
    • Simvastatin prevents oxygen and glucose deprivation/reoxygenation-induced death of cortical neurons by reducing the production and toxicity of 4-hydroxy-2E-nonenal
    • 2-s2.0-33644946838 10.1111/j.1471-4159.2006.03715.x
    • Lim J. H., Lee J.-C., Lee Y. H., Choi I. Y., Oh Y.-K., Kim H.-S., Park J.-S., Kim W.-K., Simvastatin prevents oxygen and glucose deprivation/ reoxygenation-induced death of cortical neurons by reducing the production and toxicity of 4-hydroxy-2E-nonenal. Journal of Neurochemistry 2006 97 1 140 150 2-s2.0-33644946838 10.1111/j.1471-4159.2006.03715.x
    • (2006) Journal of Neurochemistry , vol.97 , Issue.1 , pp. 140-150
    • Lim, J.H.1    Lee, J.-C.2    Lee, Y.H.3    Choi, I.Y.4    Oh, Y.-K.5    Kim, H.-S.6    Park, J.-S.7    Kim, W.-K.8
  • 249
  • 250
    • 0036088474 scopus 로고    scopus 로고
    • 4-Hydroxynonenal decreases interleukin-6 expression and protein production in primary rat Kupffer cells by inhibiting nuclear factor-κB activation
    • DOI 10.1124/jpet.102.033522
    • Luckey S. W., Taylor M., Sampey B. P., Scheinman R. I., Petersen D. R., 4-Hydroxynonenal decreases interleukin-6 expression and protein production in primary rat Kupffer cells by inhibiting nuclear factor- B activation. Journal of Pharmacology and Experimental Therapeutics 2002 302 1 296 303 2-s2.0-0036088474 10.1124/jpet.102.033522 (Pubitemid 34680451)
    • (2002) Journal of Pharmacology and Experimental Therapeutics , vol.302 , Issue.1 , pp. 296-303
    • Luckey, S.W.1    Taylor, M.2    Sampey, B.P.3    Scheinman, R.I.4    Petersen, D.R.5
  • 251
    • 0034806091 scopus 로고    scopus 로고
    • 4-Hydroxy-2-nonenal is a powerful endogenous inhibitor of endothelial response
    • DOI 10.1006/bbrc.2001.4586
    • Minekura H., Kumagai T., Kawamoto Y., Nara F., Uchida K., 4-Hydroxy-2-nonenal is a powerful endogenous inhibitor of endothelial response. Biochemical and Biophysical Research Communications 2001 282 2 557 561 2-s2.0-0034806091 10.1006/bbrc.2001.4586 (Pubitemid 32917907)
    • (2001) Biochemical and Biophysical Research Communications , vol.282 , Issue.2 , pp. 557-561
    • Minekura, H.1    Kumagai, T.2    Kawamoto, Y.3    Nara, F.4    Uchida, K.5
  • 252
    • 0035947678 scopus 로고    scopus 로고
    • I B kinase, a molecular target for inhibition by 4-hydroxy-2-nonenal
    • 2-s2.0-0035947678 10.1074/jbc.M101266200
    • Ji C., Kozak K. R., Marnett L. J., I B kinase, a molecular target for inhibition by 4-hydroxy-2-nonenal. Journal of Biological Chemistry 2001 276 21 18223 18228 2-s2.0-0035947678 10.1074/jbc.M101266200
    • (2001) Journal of Biological Chemistry , vol.276 , Issue.21 , pp. 18223-18228
    • Ji, C.1    Kozak, K.R.2    Marnett, L.J.3
  • 253
    • 77958081965 scopus 로고    scopus 로고
    • HNE-induced 5-LO expression is regulated by NF- B/ERK and Sp1/p38 MAPK pathways via EGF receptor in murine macrophages
    • 2-s2.0-77958081965 10.1093/cvr/cvq194
    • Lee S. J., Kim C. E., Seo K. W., Kim C. D., HNE-induced 5-LO expression is regulated by NF- B/ERK and Sp1/p38 MAPK pathways via EGF receptor in murine macrophages. Cardiovascular Research 2010 88 2 352 359 2-s2.0-77958081965 10.1093/cvr/cvq194
    • (2010) Cardiovascular Research , vol.88 , Issue.2 , pp. 352-359
    • Lee, S.J.1    Kim, C.E.2    Seo, K.W.3    Kim, C.D.4
  • 254
    • 54349125514 scopus 로고    scopus 로고
    • 4-hydroxynonenal enhances MMP-2 production in vascular smooth muscle cells via mitochondrial ROS-mediated activation of the Akt/NF- B signaling pathways
    • 2-s2.0-54349125514 10.1016/j.freeradbiomed.2008.08.022
    • Lee S. J., Seo K. W., Yun M. R., Bae S. S., Lee W. S., Hong K. W., Kim C. D., 4-hydroxynonenal enhances MMP-2 production in vascular smooth muscle cells via mitochondrial ROS-mediated activation of the Akt/NF- B signaling pathways. Free Radical Biology and Medicine 2008 45 10 1487 1492 2-s2.0-54349125514 10.1016/j.freeradbiomed.2008.08.022
    • (2008) Free Radical Biology and Medicine , vol.45 , Issue.10 , pp. 1487-1492
    • Lee, S.J.1    Seo, K.W.2    Yun, M.R.3    Bae, S.S.4    Lee, W.S.5    Hong, K.W.6    Kim, C.D.7
  • 255
    • 37449012959 scopus 로고    scopus 로고
    • Alterations in mitochondrial respiratory functions, redox metabolism and apoptosis by oxidant 4-hydroxynonenal and antioxidants curcumin and melatonin in PC12 cells
    • 2-s2.0-37449012959 10.1016/j.taap.2007.09.002
    • Raza H., John A., Brown E. M., Benedict S., Kambal A., Alterations in mitochondrial respiratory functions, redox metabolism and apoptosis by oxidant 4-hydroxynonenal and antioxidants curcumin and melatonin in PC12 cells. Toxicology and Applied Pharmacology 2008 226 2 161 168 2-s2.0-37449012959 10.1016/j.taap.2007.09.002
    • (2008) Toxicology and Applied Pharmacology , vol.226 , Issue.2 , pp. 161-168
    • Raza, H.1    John, A.2    Brown, E.M.3    Benedict, S.4    Kambal, A.5
  • 256
    • 33846818874 scopus 로고    scopus 로고
    • 4-Hydroxynonenal, a product of oxidative stress, leads to an antioxidant response in optic nerve head astrocytes
    • DOI 10.1016/j.exer.2006.10.020, PII S0014483506004234
    • Malone P. E., Hernandez M. R., 4-Hydroxynonenal, a product of oxidative stress, leads to an antioxidant response in optic nerve head astrocytes. Experimental Eye Research 2007 84 3 444 454 2-s2.0-33846818874 10.1016/j.exer.2006.10.020 (Pubitemid 46210058)
    • (2007) Experimental Eye Research , vol.84 , Issue.3 , pp. 444-454
    • Malone, P.E.1    Hernandez, M.R.2
  • 257
    • 33947585316 scopus 로고    scopus 로고
    • Differential regulation of cyclooxygenase-2 and inducible nitric oxide synthase by 4-hydroxynonenal in human osteoarthritic chondrocytes through ATF-2/CREB-1 transactivation and concomitant inhibition of NF-κB signaling cascade
    • DOI 10.1002/jcb.21110
    • Vaillancourt F., Morquette B., Shi Q., Fahmi H., Lavigne P., Di Battista J. A., Fernandes J. C., Benderdour M., Differential regulation of cyclooxygenase-2 and inducible nitric oxide synthase by 4-hydroxynonenal in human osteoarthritic chondrocytes through ATF-2/CREB-1 transactivation and concomitant inhibition of NF- B signaling cascade. Journal of Cellular Biochemistry 2007 100 5 1217 1231 2-s2.0-33947585316 10.1002/jcb.21110 (Pubitemid 46480404)
    • (2007) Journal of Cellular Biochemistry , vol.100 , Issue.5 , pp. 1217-1231
    • Vaillancourt, F.1    Morquette, B.2    Shi, Q.3    Fahmi, H.4    Lavigne, P.5    Di Battista, J.A.6    Fernandes, J.C.7    Benderdour, M.8
  • 258
    • 21244441080 scopus 로고    scopus 로고
    • Involvement of reactive oxygen species in cyclic stretch-induced NF-κB activation in human fibroblast cells
    • DOI 10.1038/sj.bjp.0706182
    • Amma H., Naruse K., Ishiguro N., Sokabe M., Involvement of reactive oxygen species in cyclic stretch-induced NF- B activation in human fibroblast cells. British Journal of Pharmacology 2005 145 3 364 373 2-s2.0-21244441080 10.1038/sj.bjp.0706182 (Pubitemid 40896081)
    • (2005) British Journal of Pharmacology , vol.145 , Issue.3 , pp. 364-373
    • Amma, H.1    Naruse, K.2    Ishiguro, N.3    Sokabe, M.4
  • 259
    • 18644373824 scopus 로고    scopus 로고
    • Chlamydia pneumoniae activates IKK/IκB-mediated signaling, which is inhibited by 4-HNE and following primary exposure
    • DOI 10.1016/S0021-9150(02)00198-3, PII S0021915002001983
    • Donath B., Fischer C., Page S., Prebeck S., Jilg N., Weber M., Da Costa C., Neumeier D., Miethke T., Brand K., Chlamydia pneumoniae activates IKK/I B-mediated signaling, which is inhibited by 4-HNE and following primary exposure. Atherosclerosis 2002 165 1 79 88 2-s2.0-18644373824 10.1016/S0021-9150(02)00198-3 (Pubitemid 35223170)
    • (2002) Atherosclerosis , vol.165 , Issue.1 , pp. 79-88
    • Donath, B.1    Fischer, C.2    Page, S.3    Prebeck, S.4    Jilg, N.5    Weber, M.6    Da Costa, C.7    Neumeier, D.8    Miethke, T.9    Brand, K.10
  • 260
    • 84897875039 scopus 로고    scopus 로고
    • Peroxisome-proliferator-activated receptors regulate redox signaling in the cardiovascular system
    • Kim T., Yang Q., Peroxisome-proliferator-activated receptors regulate redox signaling in the cardiovascular system. World Journal of Cardiology 2013 5 6 164 174
    • (2013) World Journal of Cardiology , vol.5 , Issue.6 , pp. 164-174
    • Kim, T.1    Yang, Q.2
  • 262
    • 47349107464 scopus 로고    scopus 로고
    • The role of PPAR ligands in controlling growth-related gene expression and their interaction with lipoperoxidation products
    • DOI 10.1155/2008/524671
    • Barrera G., Toaldo C., Pizzimenti S., Cerbone A., Pettazzoni P., Dianzani M. U., Ferretti C., The role of PPAR ligands in controlling growth-related gene expression and their interaction with lipoperoxidation products. PPAR Research 2008 2008 15 2-s2.0-47349107464 10.1155/2008/524671 524671 (Pubitemid 352000621)
    • (2008) PPAR Research , pp. 524671
    • Barrera, G.1    Toaldo, C.2    Pizzimenti, S.3    Cerbone, A.4    Pettazzoni, P.5    Dianzani, M.U.6    Ferretti, C.7
  • 263
    • 84862617186 scopus 로고    scopus 로고
    • 4-Hydroxynonenal differentially regulates adiponectin gene expression and secretion via activating PPAR γ and accelerating ubiquitin-proteasome degradation
    • 2-s2.0-81555230837 10.1016/j.mce.2011.10.027
    • Wang Z., Dou X., Gu D., Shen C., Yao T., Nguyen V., Braunschweig C., Song Z., 4-Hydroxynonenal differentially regulates adiponectin gene expression and secretion via activating PPAR γ and accelerating ubiquitin-proteasome degradation. Molecular and Cellular Endocrinology 2012 349 2 222 231 2-s2.0-81555230837 10.1016/j.mce.2011.10.027
    • (2012) Molecular and Cellular Endocrinology , vol.349 , Issue.2 , pp. 222-231
    • Wang, Z.1    Dou, X.2    Gu, D.3    Shen, C.4    Yao, T.5    Nguyen, V.6    Braunschweig, C.7    Song, Z.8
  • 264
    • 0036468883 scopus 로고    scopus 로고
    • Synergistic effect of 4-hydroxynonenal and PPAR ligands in controlling human leukemic cell growth and differentiation
    • DOI 10.1016/S0891-5849(01)00798-5, PII S0891584901007985
    • Pizzimenti S., Laurora S., Briatore F., Ferretti C., Dianzani M. U., Barrera G., Synergistic effect of 4-hydroxynonenal and PPAR ligands in controlling human leukemic cell growth and differentiation. Free Radical Biology and Medicine 2002 32 3 233 245 2-s2.0-0036468883 10.1016/S0891-5849(01)00798-5 (Pubitemid 34119402)
    • (2002) Free Radical Biology and Medicine , vol.32 , Issue.3 , pp. 233-245
    • Pizzimenti, S.1    Laurora, S.2    Briatore, F.3    Ferretti, C.4    Dianzani, M.U.5    Barrera, G.6
  • 265
    • 34247383583 scopus 로고    scopus 로고
    • 4-Hydroxynonenal and PPARγ ligands affect proliferation, differentiation, and apoptosis in colon cancer cells
    • DOI 10.1016/j.freeradbiomed.2007.02.009, PII S0891584907001104
    • Cerbone A., Toaldo C., Laurora S., Briatore F., Pizzimenti S., Dianzani M. U., Ferretti C., Barrera G., 4-hydroxynonenal and PPAR γ ligands affect proliferation, differentiation, and apoptosis in colon cancer cells. Free Radical Biology and Medicine 2007 42 11 1661 1670 2-s2.0-34247383583 10.1016/j.freeradbiomed.2007.02.009 (Pubitemid 46629012)
    • (2007) Free Radical Biology and Medicine , vol.42 , Issue.11 , pp. 1661-1670
    • Cerbone, A.1    Toaldo, C.2    Laurora, S.3    Briatore, F.4    Pizzimenti, S.5    Dianzani, M.U.6    Ferretti, C.7    Barrera, G.8
  • 266
    • 70350451479 scopus 로고    scopus 로고
    • Increased lipid oxidation causes oxidative stress, increased peroxisome proliferator-activated receptor- γ expression, and diminished pro-osteogenic Wnt signaling in the skeleton
    • 2-s2.0-70350451479 10.1074/jbc.M109.023572
    • Almeida M., Ambrogini E., Han L., Manolagas S. C., Jilka R. L., Increased lipid oxidation causes oxidative stress, increased peroxisome proliferator-activated receptor- γ expression, and diminished pro-osteogenic Wnt signaling in the skeleton. Journal of Biological Chemistry 2009 284 40 27438 27448 2-s2.0-70350451479 10.1074/jbc.M109.023572
    • (2009) Journal of Biological Chemistry , vol.284 , Issue.40 , pp. 27438-27448
    • Almeida, M.1    Ambrogini, E.2    Han, L.3    Manolagas, S.C.4    Jilka, R.L.5
  • 267
    • 33947241783 scopus 로고    scopus 로고
    • The oxidative stress mediator 4-hydroxynonenal is an intracellular agonist of the nuclear receptor peroxisome proliferator-activated receptor-β/δ (PPARβ/δ)
    • DOI 10.1016/j.freeradbiomed.2007.01.003, PII S089158490700010X
    • Coleman J. D., Prabhu K. S., Thompson J. T., Reddy P. S., Peters J. M., Peterson B. R., Reddy C. C., Vanden Heuvel J. P., The oxidative stress mediator 4-hydroxynonenal is an intracellular agonist of the nuclear receptor peroxisome proliferator-activated receptor- β / δ (PPAR β / δ). Free Radical Biology and Medicine 2007 42 8 1155 1164 2-s2.0-33947241783 10.1016/j.freeradbiomed.2007.01.003 (Pubitemid 46428510)
    • (2007) Free Radical Biology and Medicine , vol.42 , Issue.8 , pp. 1155-1164
    • Coleman, J.D.1    Prabhu, K.S.2    Thompson, J.T.3    Reddy, P.S.4    Peters, J.M.5    Peterson, B.R.6    Reddy, C.C.7    Vanden Heuvel, J.P.8
  • 268
  • 270
    • 0041669519 scopus 로고    scopus 로고
    • 4-Hydroxy-2-nonenal as a COX-2 inducer
    • DOI 10.1016/S0098-2997(03)00016-5
    • Uchida K., Kumagai T., 4-Hydroxy-2-nonenal as a COX-2 inducer. Molecular Aspects of Medicine 2003 24 4-5 213 218 2-s2.0-0041669519 10.1016/S0098-2997(03) 00016-5 (Pubitemid 36945021)
    • (2003) Molecular Aspects of Medicine , vol.24 , Issue.4-5 , pp. 213-218
    • Uchida, K.1    Kumagai, T.2
  • 272
    • 45849089850 scopus 로고    scopus 로고
    • SHP-1 inhibition by 4-hydroxynonenal activates Jun N-terminal kinase and glutamate cysteine ligase
    • DOI 10.1165/rcmb.2007-0371OC
    • Rinna A., Forman H. J., SHP-1 inhibition by 4-hydroxynonenal activates Jun N-terminal kinase and glutamate cysteine ligase. American Journal of Respiratory Cell and Molecular Biology 2008 39 1 97 104 2-s2.0-45849089850 10.1165/rcmb.2007-0371OC (Pubitemid 351883963)
    • (2008) American Journal of Respiratory Cell and Molecular Biology , vol.39 , Issue.1 , pp. 97-104
    • Rinna, A.1    Forman, H.J.2
  • 273
    • 0035047707 scopus 로고    scopus 로고
    • 4-Hydroxy-2-nonenal increases γ-glutamylcysteine synthetase gene expression in alveolar epithelial cells
    • Liu R.-M., Borok Z., Forman H. J., 4-Hydroxy-2-nonenal increases γ -glutamylcysteine synthetase gene expression in alveolar epithelial cells. American Journal of Respiratory Cell and Molecular Biology 2001 24 4 499 505 2-s2.0-0035047707 (Pubitemid 32324740)
    • (2001) American Journal of Respiratory Cell and Molecular Biology , vol.24 , Issue.4 , pp. 499-505
    • Liu, R.-M.1    Borok, Z.2    Forman, H.J.3
  • 274
    • 0036802703 scopus 로고    scopus 로고
    • 4-Hydroxynonenal induces glutamate cysteine ligase through JNK in HBE1 cells
    • DOI 10.1016/S0891-5849(02)00991-7, PII S0891584902009917
    • Dickinson D. A., Iles K. E., Watanabe N., Iwamoto T., Zhang H., Krzywanski D. M., Forman H. J., 4-Hydroxynonenal induces glutamate cysteine ligase through JNK in HBE1 cells. Free Radical Biology and Medicine 2002 33 7 974 987 2-s2.0-0036802703 10.1016/S0891-5849(02)00991-7 (Pubitemid 35284035)
    • (2002) Free Radical Biology and Medicine , vol.33 , Issue.7 , pp. 974-987
    • Dickinson, D.A.1    Iles, K.E.2    Watanabe, N.3    Iwamoto, T.4    Zhang, H.5    Krzywanski, D.M.6    Forman, H.J.7
  • 275
    • 53149101293 scopus 로고    scopus 로고
    • Inhibition of the lipopolysaccharide-induced stimulation of the members of the MAPK family in human monocytes/macrophages by 4-hydroxynonenal, a product of oxidized omega-6 fatty acids
    • 2-s2.0-53149101293 10.2353/ajpath.2008.071150
    • Marantos C., Mukaro V., Ferrante J., Hii C., Ferrante A., Inhibition of the lipopolysaccharide-induced stimulation of the members of the MAPK family in human monocytes/macrophages by 4-hydroxynonenal, a product of oxidized omega-6 fatty acids. American Journal of Pathology 2008 173 4 1057 1066 2-s2.0-53149101293 10.2353/ajpath.2008.071150
    • (2008) American Journal of Pathology , vol.173 , Issue.4 , pp. 1057-1066
    • Marantos, C.1    Mukaro, V.2    Ferrante, J.3    Hii, C.4    Ferrante, A.5
  • 276
  • 277
    • 33845629703 scopus 로고    scopus 로고
    • Redox regulation of 4-hydroxy-2-nonenal-mediated endothelial barrier dysfunction by focal adhesion, adherens, and tight junction proteins
    • DOI 10.1074/jbc.M607305200
    • Usatyuk P. V., Parinandi N. L., Natarajan V., Redox regulation of 4-hydroxy-2-nonenal-mediated endothelial barrier dysfunction by focal adhesion, adherens, and tight junction proteins. Journal of Biological Chemistry 2006 281 46 35554 35566 2-s2.0-33845629703 10.1074/jbc.M607305200 (Pubitemid 46036589)
    • (2006) Journal of Biological Chemistry , vol.281 , Issue.46 , pp. 35554-35566
    • Usatyuk, P.V.1    Parinandi, N.L.2    Natarajan, V.3
  • 279
    • 84878109046 scopus 로고    scopus 로고
    • Matrix metalloproteinase-2 and -9 as promising benefactors in development, plasticity and repair of the nervous system
    • Verslegers M., Lemmens K., Van Hove I., Moons L., Matrix metalloproteinase-2 and -9 as promising benefactors in development, plasticity and repair of the nervous system. Progress in Neurobiology 2013 105 60 78
    • (2013) Progress in Neurobiology , vol.105 , pp. 60-78
    • Verslegers, M.1    Lemmens, K.2    Van Hove, I.3    Moons, L.4
  • 280
    • 71049193819 scopus 로고    scopus 로고
    • 4-Hydroxynonenal enhances MMP-9 production in murine macrophages via 5-lipoxygenase-mediated activation of ERK and p38 MAPK
    • 2-s2.0-71049193819 10.1016/j.taap.2009.10.007
    • Lee S. J., Kim C. E., Yun M. R., Seo K. W., Park H. M., Yun J. W., Shin H. K., Bae S. S., Kim C. D., 4-Hydroxynonenal enhances MMP-9 production in murine macrophages via 5-lipoxygenase-mediated activation of ERK and p38 MAPK. Toxicology and Applied Pharmacology 2010 242 2 191 198 2-s2.0-71049193819 10.1016/j.taap.2009.10.007
    • (2010) Toxicology and Applied Pharmacology , vol.242 , Issue.2 , pp. 191-198
    • Lee, S.J.1    Kim, C.E.2    Yun, M.R.3    Seo, K.W.4    Park, H.M.5    Yun, J.W.6    Shin, H.K.7    Bae, S.S.8    Kim, C.D.9
  • 281
    • 73449139739 scopus 로고    scopus 로고
    • Participation of 5-lipoxygenase-derived LTB4 in 4-hydroxynonenal-enhanced MMP-2 production in vascular smooth muscle cells
    • 2-s2.0-73449139739 10.1016/j.atherosclerosis.2009.06.012
    • Seo K. W., Lee S. J., Kim C. E., Yun M. R., Park H. M., Yun J. W., Bae S. S., Kim C. D., Participation of 5-lipoxygenase-derived LTB4 in 4-hydroxynonenal-enhanced MMP-2 production in vascular smooth muscle cells. Atherosclerosis 2010 208 1 56 61 2-s2.0-73449139739 10.1016/j.atherosclerosis. 2009.06.012
    • (2010) Atherosclerosis , vol.208 , Issue.1 , pp. 56-61
    • Seo, K.W.1    Lee, S.J.2    Kim, C.E.3    Yun, M.R.4    Park, H.M.5    Yun, J.W.6    Bae, S.S.7    Kim, C.D.8
  • 282
    • 31044435908 scopus 로고    scopus 로고
    • Production of lipid peroxidation products in osteoarthritic tissues: New evidence linking 4-hydroxynonenal to cartilage degradation
    • DOI 10.1002/art.21559
    • Morquette B., Shi Q., Lavigne P., Ranger P., Fernandes J. C., Benderdour M., Production of lipid peroxidation products in osteoarthritic tissues: new evidence linking 4-hydroxynonenal to cartilage degradation. Arthritis and Rheumatism 2006 54 1 271 281 2-s2.0-31044435908 10.1002/art.21559 (Pubitemid 43122212)
    • (2006) Arthritis and Rheumatism , vol.54 , Issue.1 , pp. 271-281
    • Morquette, B.1    Shi, Q.2    Lavigne, P.3    Ranger, P.4    Fernandes, J.C.5    Benderdour, M.6
  • 283
    • 79960716001 scopus 로고    scopus 로고
    • Akt signalling in health and disease
    • Hers I., Vincent E. E., Tavaré J. M., Akt signalling in health and disease. Cell Signaling 2011 23 10 1515 1527
    • (2011) Cell Signaling , vol.23 , Issue.10 , pp. 1515-1527
    • Hers, I.1    Vincent, E.E.2    Tavaré, J.M.3
  • 285
    • 79955830382 scopus 로고    scopus 로고
    • Modification of Akt2 by 4-hydroxynonenal inhibits insulin-dependent Akt signaling in HepG2 cells
    • 2-s2.0-79955830382 10.1021/bi200029w
    • Shearn C. T., Fritz K. S., Reigan P., Petersen D. R., Modification of Akt2 by 4-hydroxynonenal inhibits insulin-dependent Akt signaling in HepG2 cells. Biochemistry 2011 50 19 3984 3996 2-s2.0-79955830382 10.1021/bi200029w
    • (2011) Biochemistry , vol.50 , Issue.19 , pp. 3984-3996
    • Shearn, C.T.1    Fritz, K.S.2    Reigan, P.3    Petersen, D.R.4
  • 286
    • 84883177695 scopus 로고    scopus 로고
    • Increased carbonylation of the lipid phosphatase PTEN contributes to Akt2 activation in a murine model of early alcohol-induced steatosis
    • Shearn C. T., Smathers R. L., Backos D. S., Reigan P., Orlicky D. J., Petersen D. R., Increased carbonylation of the lipid phosphatase PTEN contributes to Akt2 activation in a murine model of early alcohol-induced steatosis. Free Radical Biology and Medicine 2013 65 680 692
    • (2013) Free Radical Biology and Medicine , vol.65 , pp. 680-692
    • Shearn, C.T.1    Smathers, R.L.2    Backos, D.S.3    Reigan, P.4    Orlicky, D.J.5    Petersen, D.R.6
  • 287
    • 84861370262 scopus 로고    scopus 로고
    • Inhibition of Hydrogen peroxide signaling by 4-hydroxynonenal due to differential regulation of Akt1 and Akt2 contributes to decreases in cell survival and proliferation in hepatocellular carcinoma cells
    • Shearn C. T., Reigan P., Petersen D. R., Inhibition of Hydrogen peroxide signaling by 4-hydroxynonenal due to differential regulation of Akt1 and Akt2 contributes to decreases in cell survival and proliferation in hepatocellular carcinoma cells. Free Radical Biology and Medicine 2012 53 1 1 11
    • (2012) Free Radical Biology and Medicine , vol.53 , Issue.1 , pp. 1-11
    • Shearn, C.T.1    Reigan, P.2    Petersen, D.R.3
  • 288
    • 79451476147 scopus 로고    scopus 로고
    • Role of 4-hydroxynonenal in epidermal growth factor receptor-mediated signaling in retinal pigment epithelial cells
    • 2-s2.0-79451476147 10.1016/j.exer.2010.11.010
    • Vatsyayan R., Chaudhary P., Sharma A., Sharma R., Rao Lelsani P. C., Awasthi S., Awasthi Y. C., Role of 4-hydroxynonenal in epidermal growth factor receptor-mediated signaling in retinal pigment epithelial cells. Experimental Eye Research 2011 92 2 147 154 2-s2.0-79451476147 10.1016/j.exer.2010.11.010
    • (2011) Experimental Eye Research , vol.92 , Issue.2 , pp. 147-154
    • Vatsyayan, R.1    Chaudhary, P.2    Sharma, A.3    Sharma, R.4    Rao Lelsani, P.C.5    Awasthi, S.6    Awasthi, Y.C.7
  • 289
    • 78651376861 scopus 로고    scopus 로고
    • Protein kinase C isoforms: Mediators of reactive lipid metabolites in the development of insulin resistance
    • 2-s2.0-78651376861 10.1016/j.febslet.2010.12.022
    • Turban S., Hajduch E., Protein kinase C isoforms: mediators of reactive lipid metabolites in the development of insulin resistance. FEBS Letters 2011 585 2 269 274 2-s2.0-78651376861 10.1016/j.febslet.2010.12.022
    • (2011) FEBS Letters , vol.585 , Issue.2 , pp. 269-274
    • Turban, S.1    Hajduch, E.2
  • 290
    • 31644440260 scopus 로고    scopus 로고
    • The exocytosis induced in HL-60 cells by 4-hydroxynonenal, a lipid peroxidation product, is not prevented by reduced glutathione
    • DOI 10.1002/cbf.1260
    • Maggiora M., Rossi M. A., The exocytosis induced in HL-60 cells by 4-hydroxynonenal, a lipid peroxidation product, is not prevented by reduced glutathione. Cell Biochemistry and Function 2006 24 1 1 6 2-s2.0-31644440260 10.1002/cbf.1260 (Pubitemid 43171002)
    • (2006) Cell Biochemistry and Function , vol.24 , Issue.1 , pp. 1-6
    • Maggiora, M.1    Rossi, M.A.2
  • 291
    • 0037911448 scopus 로고    scopus 로고
    • Experimental researches on the role of phosphoinositide-specific phospholipase C in 4-hydroxynonenal induced exocytosis
    • DOI 10.1002/cbf.1013
    • Maggiora M., Rossi M. A., Experimental researches on the role of phosphoinositide-specific phospholipase C in 4-hydroxynonenal induced exocytosis. Cell Biochemistry and Function 2003 21 2 155 160 2-s2.0-0037911448 10.1002/cbf.1013 (Pubitemid 36597183)
    • (2003) Cell Biochemistry and Function , vol.21 , Issue.2 , pp. 155-160
    • Maggiora, M.1    Rossi, M.A.2
  • 292
    • 0034935945 scopus 로고    scopus 로고
    • Experimental studies on the mechanism of phospholipase C activation by the lipid peroxidation products 4-hydroxynonenal and 2-nonenal
    • Rossi M. A., Di Mauro C., Dianzani M. U., Experimental studies on the mechanism of phospholipase C activation by the lipid peroxidation products 4-hydroxynonenal and 2-nonenal. International Journal of Tissue Reactions 2001 23 2 45 50 2-s2.0-0034935945 (Pubitemid 32618842)
    • (2001) International Journal of Tissue Reactions , vol.23 , Issue.2 , pp. 45-50
    • Rossi, M.A.1    Di Mauro, C.2    Dianzani, M.U.3
  • 293
    • 0028005464 scopus 로고
    • Activation of phosphoinositide-specific phospholipase C of rat neutrophils by the chemotactic aldehydes 4-hydroxy-2,3-trans-nonenal and 4-hydroxy-2,3-trans-octenal
    • DOI 10.1002/cbf.290120408
    • Rossi M. A., Di Mauro C., Esterbauer H., Fidale F., Dianzani M. U., Activation of phosphoinositide-specific phospholipase C of rat neutrophils by the chemotactic aldehydes 4-hydroxy-2,3-trans-nonenal and 4-hydroxy-2,3-trans- octenal. Cell Biochemistry and Function 1994 12 4 275 280 2-s2.0-0028005464 10.1002/cbf.290120408 (Pubitemid 24350569)
    • (1994) Cell Biochemistry and Function , vol.12 , Issue.4 , pp. 275-280
    • Rossi, M.A.1    Di Mauro, C.2    Esterbauer, H.3    Fidale, F.4    Dianzani, M.U.5
  • 294
    • 79953031472 scopus 로고    scopus 로고
    • The human NADPH oxidase: Primary and secondary defects impairing the respiratory burst function and the microbicidal ability of phagocytes
    • 2-s2.0-79953031472 10.1111/j.1365-3083.2010.02501.x
    • de Oliveira-Junior E. B., Bustamante J., Newburger P. E., Condino-Neto A., The human NADPH oxidase: primary and secondary defects impairing the respiratory burst function and the microbicidal ability of phagocytes. Scandinavian Journal of Immunology 2011 73 5 420 427 2-s2.0-79953031472 10.1111/j.1365-3083.2010.02501.x
    • (2011) Scandinavian Journal of Immunology , vol.73 , Issue.5 , pp. 420-427
    • De Oliveira-Junior, E.B.1    Bustamante, J.2    Newburger, P.E.3    Condino-Neto, A.4
  • 296
    • 17044441984 scopus 로고    scopus 로고
    • Regulation of rat hepatocyte protein kinase C β isoenzymes by the lipid peroxidation product 4-hydroxy-2,3-nonenal: A signaling pathway to modulate vesicular transport of glycoproteins
    • DOI 10.1002/hep.510290510
    • Chiarpotto E., Domenicotti C., Paola D., Vitali A., Nitti M., Pronzato M. A., Biasi F., Cottalasso D., Marinari U. M., Dragonetti A., Cesaro P., Isidoro C., Poli G., Regulation of rat hepatocyte protein kinase C β isoenzymes by the lipid peroxidation product 4-hydroxy-2,3-nonenal: a signaling pathway to modulate vesicular transport of glycoproteins. Hepatology 1999 29 5 1565 1572 2-s2.0-17044441984 10.1002/hep.510290510 (Pubitemid 29211062)
    • (1999) Hepatology , vol.29 , Issue.5 , pp. 1565-1572
    • Chiarpotto, E.1
  • 298
    • 0043172499 scopus 로고    scopus 로고
    • Role of PKC-dependent pathways in HNE-induced cell protein transport and secretion
    • DOI 10.1016/S0098-2997(03)00015-3
    • Marinari U. M., Nitti M., Pronzato M. A., Domenicotti C., Role of PKC-dependent pathways in HNE-induced cell protein transport and secretion. Molecular Aspects of Medicine 2003 24 4-5 205 211 2-s2.0-0043172499 10.1016/S0098-2997(03)00015-3 (Pubitemid 36945020)
    • (2003) Molecular Aspects of Medicine , vol.24 , Issue.4-5 , pp. 205-211
    • Marinari, U.M.1    Nitti, M.2    Pronzato, M.A.3    Domenicotti, C.4
  • 300
    • 33845962827 scopus 로고    scopus 로고
    • Aldose reductase mediates the lipopolysaccharide-induced release of inflammatory mediators in RAW264.7 murine macrophages
    • DOI 10.1074/jbc.M603819200
    • Ramana K. V., Fadl A. A., Tammali R., Reddy A. B. M., Chopra A. K., Srivastava S. K., Aldose reductase mediates the lipopolysaccharide-induced release of inflammatory mediators in RAW264.7 murine macrophages. Journal of Biological Chemistry 2006 281 44 33019 33029 2-s2.0-33845962827 10.1074/jbc.M603819200 (Pubitemid 46036684)
    • (2006) Journal of Biological Chemistry , vol.281 , Issue.44 , pp. 33019-33029
    • Ramana, K.V.1    Fadl, A.A.2    Tammali, R.3    Reddy, A.B.M.4    Chopra, A.K.5    Srivastava, S.K.6
  • 301
    • 84879047011 scopus 로고    scopus 로고
    • Cellular metabolic and autophagic pathways: Traffic control by redox signaling
    • Dodson M., Darley-Usmar V., Zhang J., Cellular metabolic and autophagic pathways: traffic control by redox signaling. Free Radical Biology and Medicine 2013 63 207 221
    • (2013) Free Radical Biology and Medicine , vol.63 , pp. 207-221
    • Dodson, M.1    Darley-Usmar, V.2    Zhang, J.3
  • 302
    • 41149116150 scopus 로고    scopus 로고
    • Unsaturated lipid peroxidation-derived aldehydes activate autophagy in vascular smooth-muscle cells
    • DOI 10.1042/BJ20071063
    • Hill B. G., Haberzettl P., Ahmed Y., Srivastava S., Bhatnagar A., Unsaturated lipid peroxidation-derived aldehydes activate autophagy in vascular smooth-muscle cells. Biochemical Journal 2008 410 3 525 534 2-s2.0-41149116150 10.1042/BJ20071063 (Pubitemid 351429023)
    • (2008) Biochemical Journal , vol.410 , Issue.3 , pp. 525-534
    • Hill, B.G.1    Haberzettl, P.2    Ahmed, Y.3    Srivastava, S.4    Bhatnagar, A.5
  • 303
    • 84878772042 scopus 로고    scopus 로고
    • Oxidized lipids activate autophagy in a JNK-dependent manner by stimulating the endoplasmic reticulum stress response
    • Haberzettl P., Hill B. G., Oxidized lipids activate autophagy in a JNK-dependent manner by stimulating the endoplasmic reticulum stress response. Redox Biology 2013 1 1 56 64
    • (2013) Redox Biology , vol.1 , Issue.1 , pp. 56-64
    • Haberzettl, P.1    Hill, B.G.2
  • 304
    • 84890822456 scopus 로고    scopus 로고
    • Inhibition of glycolysis attenuates 4-hydroxynonenal-dependent autophagy and exacerbates apoptosis in differentiated SH-SY5Y neuroblastoma cells
    • Dodson M., Liang Q., Johnson M. S., Redmann M., Fineberg N., Darley-Usmar V. M., Zhang J., Inhibition of glycolysis attenuates 4-hydroxynonenal-dependent autophagy and exacerbates apoptosis in differentiated SH-SY5Y neuroblastoma cells. Autophagy 2013 9 12 1996 2008
    • (2013) Autophagy , vol.9 , Issue.12 , pp. 1996-2008
    • Dodson, M.1    Liang, Q.2    Johnson, M.S.3    Redmann, M.4    Fineberg, N.5    Darley-Usmar, V.M.6    Zhang, J.7
  • 305
    • 77649188326 scopus 로고    scopus 로고
    • Effects of lipid peroxidation products on lipofuscinogenesis and autophagy in human retinal pigment epithelial cells
    • 2-s2.0-77649188326 10.1016/j.exer.2009.12.011
    • Krohne T. U., Stratmann N. K., Kopitz J., Holz F. G., Effects of lipid peroxidation products on lipofuscinogenesis and autophagy in human retinal pigment epithelial cells. Experimental Eye Research 2010 90 3 465 471 2-s2.0-77649188326 10.1016/j.exer.2009.12.011
    • (2010) Experimental Eye Research , vol.90 , Issue.3 , pp. 465-471
    • Krohne, T.U.1    Stratmann, N.K.2    Kopitz, J.3    Holz, F.G.4
  • 306
    • 84876912049 scopus 로고    scopus 로고
    • The roles of senescence and telomere shortening in cardiovascular disease
    • Fyhrquist F., Saijonmaa O., Strandberg T., The roles of senescence and telomere shortening in cardiovascular disease. Nature Reviews Cardiology 2013 10 5 274 283
    • (2013) Nature Reviews Cardiology , vol.10 , Issue.5 , pp. 274-283
    • Fyhrquist, F.1    Saijonmaa, O.2    Strandberg, T.3
  • 307
    • 77951977367 scopus 로고    scopus 로고
    • Endogenous DNA breaks: GammaH2AX and the role of telomeres
    • 2-s2.0-77951977367
    • Olive P. L., Endogenous DNA breaks: gammaH2AX and the role of telomeres. Aging 2009 1 2 154 156 2-s2.0-77951977367
    • (2009) Aging , vol.1 , Issue.2 , pp. 154-156
    • Olive, P.L.1
  • 308
    • 84873319024 scopus 로고    scopus 로고
    • The role of telomeres in stem cells and cancer
    • Günes C., Rudolph K. L., The role of telomeres in stem cells and cancer. Cell 2013 152 3 390 393
    • (2013) Cell , vol.152 , Issue.3 , pp. 390-393
    • Günes, C.1    Rudolph, K.L.2
  • 309
    • 67349218427 scopus 로고    scopus 로고
    • Adduct formation of 4-hydroxynonenal and malondialdehyde with elongation factor-2 in vitro and in vivo
    • 2-s2.0-67349218427 10.1016/j.freeradbiomed.2009.05.010
    • Argüelles S., Machado A., Ayala A., Adduct formation of 4-hydroxynonenal and malondialdehyde with elongation factor-2 in vitro and in vivo. Free Radical Biology and Medicine 2009 47 3 324 330 2-s2.0-67349218427 10.1016/j.freeradbiomed.2009.05.010
    • (2009) Free Radical Biology and Medicine , vol.47 , Issue.3 , pp. 324-330
    • Argüelles, S.1    Machado, A.2    Ayala, A.3
  • 311
    • 84862794254 scopus 로고    scopus 로고
    • A senescent cell bystander effect: Senescence-induced senescence
    • 2-s2.0-84858343297 10.1111/j.1474-9726.2012.00795.x
    • Nelson G., Wordsworth J., Wang C., Jurk D., Lawless C., Martin-Ruiz C., von Zglinicki T., A senescent cell bystander effect: senescence-induced senescence. Aging Cell 2012 11 2 345 349 2-s2.0-84858343297 10.1111/j.1474-9726. 2012.00795.x
    • (2012) Aging Cell , vol.11 , Issue.2 , pp. 345-349
    • Nelson, G.1    Wordsworth, J.2    Wang, C.3    Jurk, D.4    Lawless, C.5    Martin-Ruiz, C.6    Von Zglinicki, T.7
  • 312
    • 41449094458 scopus 로고    scopus 로고
    • Chronic treatment with N-acetyl-cystein delays cellular senescence in endothelial cells isolated from a subgroup of atherosclerotic patients
    • 2-s2.0-41449094458 10.1016/j.mad.2008.01.004
    • Voghel G., Thorin-Trescases N., Farhat N., Mamarbachi A. M., Villeneuve L., Fortier A., Perrault L. P., Carrier M., Thorin E., Chronic treatment with N-acetyl-cystein delays cellular senescence in endothelial cells isolated from a subgroup of atherosclerotic patients. Mechanisms of Ageing and Development 2008 129 5 261 270 2-s2.0-41449094458 10.1016/j.mad.2008.01.004
    • (2008) Mechanisms of Ageing and Development , vol.129 , Issue.5 , pp. 261-270
    • Voghel, G.1    Thorin-Trescases, N.2    Farhat, N.3    Mamarbachi, A.M.4    Villeneuve, L.5    Fortier, A.6    Perrault, L.P.7    Carrier, M.8    Thorin, E.9
  • 314
    • 77955843950 scopus 로고    scopus 로고
    • 4-Hydroxynonenal, a lipid peroxidation product of dietary polyunsaturated fatty acids, has anticarcinogenic properties in colon carcinoma cell lines through the inhibition of telomerase activity
    • 2-s2.0-77955843950 10.1016/j.jnutbio.2009.06.005
    • Pizzimenti S., Menegatti E., Berardi D., Toaldo C., Pettazzoni P., Minelli R., Giglioni B., Cerbone A., Dianzani M. U., Ferretti C., Barrera G., 4-Hydroxynonenal, a lipid peroxidation product of dietary polyunsaturated fatty acids, has anticarcinogenic properties in colon carcinoma cell lines through the inhibition of telomerase activity. Journal of Nutritional Biochemistry 2010 21 9 818 826 2-s2.0-77955843950 10.1016/j.jnutbio.2009.06.005
    • (2010) Journal of Nutritional Biochemistry , vol.21 , Issue.9 , pp. 818-826
    • Pizzimenti, S.1    Menegatti, E.2    Berardi, D.3    Toaldo, C.4    Pettazzoni, P.5    Minelli, R.6    Giglioni, B.7    Cerbone, A.8    Dianzani, M.U.9    Ferretti, C.10    Barrera, G.11
  • 315
    • 84886794616 scopus 로고    scopus 로고
    • Senescence and aging: The critical roles of p53
    • Rufini A., Tucci P., Celardo I., Melino G., Senescence and aging: the critical roles of p53. Oncogene 2013 32 43 5129 5143
    • (2013) Oncogene , vol.32 , Issue.43 , pp. 5129-5143
    • Rufini, A.1    Tucci, P.2    Celardo, I.3    Melino, G.4
  • 316
    • 84878862873 scopus 로고    scopus 로고
    • Senescence regulation by the p53 protein family
    • Qian Y., Chen X., Senescence regulation by the p53 protein family. Methods in Molecular Biology 2013 965 37 61
    • (2013) Methods in Molecular Biology , vol.965 , pp. 37-61
    • Qian, Y.1    Chen, X.2
  • 318
    • 79961180595 scopus 로고    scopus 로고
    • P53, oxidative stress, and aging
    • 2-s2.0-79961180595 10.1089/ars.2010.3644
    • Liu D., Xu Y., P53, oxidative stress, and aging. Antioxidants and Redox Signaling 2011 15 6 1669 1678 2-s2.0-79961180595 10.1089/ars.2010.3644
    • (2011) Antioxidants and Redox Signaling , vol.15 , Issue.6 , pp. 1669-1678
    • Liu, D.1    Xu, Y.2
  • 319
    • 79961199862 scopus 로고    scopus 로고
    • Redox control and interplay between p53 isoforms: Roles in the regulation of basal p53 levels, cell fate, and senescence
    • 2-s2.0-79961199862 10.1089/ars.2010.3771
    • Hafsi H., Hainaut P., Redox control and interplay between p53 isoforms: roles in the regulation of basal p53 levels, cell fate, and senescence. Antioxidants and Redox Signaling 2011 15 6 1655 1667 2-s2.0-79961199862 10.1089/ars.2010.3771
    • (2011) Antioxidants and Redox Signaling , vol.15 , Issue.6 , pp. 1655-1667
    • Hafsi, H.1    Hainaut, P.2
  • 320
    • 78650152988 scopus 로고    scopus 로고
    • P53, ROS and senescence in the control of aging
    • 2-s2.0-78650152988
    • Vigneron A., Vousden K. H., p53, ROS and senescence in the control of aging. Aging 2010 2 8 471 474 2-s2.0-78650152988
    • (2010) Aging , vol.2 , Issue.8 , pp. 471-474
    • Vigneron, A.1    Vousden, K.H.2
  • 321
    • 84867659145 scopus 로고    scopus 로고
    • The influence of reactive oxygen species on cell cycle progression in mammalian cells
    • Verbon E. H., Post J. A., Boonstra J., The influence of reactive oxygen species on cell cycle progression in mammalian cells. Gene 2012 511 1 1 6
    • (2012) Gene , vol.511 , Issue.1 , pp. 1-6
    • Verbon, E.H.1    Post, J.A.2    Boonstra, J.3
  • 322
    • 84867871087 scopus 로고    scopus 로고
    • Redox control of cell proliferation
    • Chiu J., Dawes I. W., Redox control of cell proliferation. Trends in Cell Biology 2012 22 11 592 601
    • (2012) Trends in Cell Biology , vol.22 , Issue.11 , pp. 592-601
    • Chiu, J.1    Dawes, I.W.2
  • 323
    • 84880264045 scopus 로고    scopus 로고
    • Cdks, cyclins and CKIs: Roles beyond cell cycle regulation
    • Lim S., Kaldis P., Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development 2013 140 15 3079 3093
    • (2013) Development , vol.140 , Issue.15 , pp. 3079-3093
    • Lim, S.1    Kaldis, P.2
  • 325
    • 0033004490 scopus 로고    scopus 로고
    • Inhibition of D1, D2, and A cyclin expression in HL-60 cells by the lipid peroxydation product 4-hydroxynonenal
    • DOI 10.1016/S0891-5849(99)00022-2, PII S0891584999000222
    • Pizzimenti S., Barrera G., Dianzani M. U., Brüsselbach S., Inhibition of D1, D2, and A cyclin expression in HL-60 cells by the lipid peroxydation product 4-hydroxynonenal. Free Radical Biology and Medicine 1999 26 11-12 1578 1586 2-s2.0-0033004490 10.1016/S0891-5849(99)00022-2 (Pubitemid 29302291)
    • (1999) Free Radical Biology and Medicine , vol.26 , Issue.11-12 , pp. 1578-1586
    • Pizzimenti, S.1    Barrera, G.2    Dianzani, M.U.3    Brusselbach, S.4
  • 326
    • 78549286042 scopus 로고    scopus 로고
    • Inhibition of erythropoiesis in malaria anemia: Role of hemozoin and hemozoin-generated 4-hydroxynonenal
    • 2-s2.0-78549286042 10.1182/blood-2010-03-272781
    • Skorokhod O. A., Caione L., Marrocco T., Migliardi G., Barrera V., Arese P., Piacibello W., Schwarzer E., Inhibition of erythropoiesis in malaria anemia: role of hemozoin and hemozoin-generated 4-hydroxynonenal. Blood 2010 116 20 4328 4337 2-s2.0-78549286042 10.1182/blood-2010-03-272781
    • (2010) Blood , vol.116 , Issue.20 , pp. 4328-4337
    • Skorokhod, O.A.1    Caione, L.2    Marrocco, T.3    Migliardi, G.4    Barrera, V.5    Arese, P.6    Piacibello, W.7    Schwarzer, E.8
  • 327
    • 24344490190 scopus 로고    scopus 로고
    • Breast cancer cell-targeted oxidative stress: Enhancement of cancer cell uptake of conjugated linoleic acid, activation of p53, and inhibition of proliferation
    • DOI 10.1016/j.yexmp.2005.05.005, PII S0014480005000663
    • Albright C. D., Klem E., Shah A. A., Gallagher P., Breast cancer cell-targeted oxidative stress: enhancement of cancer cell uptake of conjugated linoleic acid, activation of p53, and inhibition of proliferation. Experimental and Molecular Pathology 2005 79 2 118 125 2-s2.0-24344490190 10.1016/j.yexmp.2005.05.005 (Pubitemid 41262493)
    • (2005) Experimental and Molecular Pathology , vol.79 , Issue.2 , pp. 118-125
    • Albright, C.D.1    Klem, E.2    Shah, A.A.3    Gallagher, P.4
  • 328
    • 28644440011 scopus 로고    scopus 로고
    • The influence of 4-hydroxy-2-nonenal on proliferation, differentiation and apoptosis of human osteosarcoma cells
    • HNE and Further Lipid Peroxidation Products
    • Sunjic S. B., Cipak A., Rabuzin F., Wildburger R., Zarkovic N., The influence of 4-hydroxy-2-nonenal on proliferation, differentiation and apoptosis of human osteosarcoma cells. BioFactors 2005 24 1-4 141 148 2-s2.0-28644440011 (Pubitemid 41750537)
    • (2005) BioFactors , vol.24 , Issue.1-4 , pp. 141-148
    • Sunjic, S.B.1    Cipak, A.2    Rabuzin, F.3    Wildburger, R.4    Zarkovic, N.5
  • 329
    • 0037324844 scopus 로고    scopus 로고
    • Antisense oligonucleotides against aldehyde dehydrogenase 3 inhibit hepatoma cell proliferation by affecting MAP kinases
    • DOI 10.1016/S0009-2797(02)00170-9, PII S0009279702001709
    • Muzio G., Trombetta A., Martinasso G., Canuto R. A., Maggiora M., Antisense oligonucleotides against aldehyde dehydrogenase 3 inhibit hepatoma cell proliferation by affecting MAP kinases. Chemico-Biological Interactions 2003 143-144 37 43 2-s2.0-0037324844 10.1016/S0009-2797(02)00170-9 (Pubitemid 36253577)
    • (2003) Chemico-Biological Interactions , vol.143-144 , pp. 37-43
    • Muzio, G.1    Trombetta, A.2    Martinasso, G.3    Canuto, R.A.4    Maggiora, M.5
  • 330
    • 0032957410 scopus 로고    scopus 로고
    • Inhibition of class-3 aldehyde dehydrogenase and cell growth by restored lipid peroxidation in hepatoma cell lines
    • DOI 10.1016/S0891-5849(98)00206-8, PII S0891584998002068
    • Canuto R. A., Muzio G., Ferro M., Maggiora M., Federa R., Bassi A. M., Lindahl R., Dianzani M. U., Inhibition of class-3 aldehyde dehydrogenase and cell growth by restored lipid peroxidation in hepatoma cell lines. Free Radical Biology and Medicine 1999 26 3-4 333 340 2-s2.0-0032957410 10.1016/S0891- 5849(98)00206-8 (Pubitemid 29013464)
    • (1999) Free Radical Biology and Medicine , vol.26 , Issue.3-4 , pp. 333-340
    • Canuto, R.A.1    Muzio, G.2    Ferro, M.3    Maggiora, M.4    Federa, R.5    Bassi, A.M.6    Lindahl, R.7    Dianzani, M.U.8
  • 331
    • 57349111927 scopus 로고    scopus 로고
    • Down-regulation of Notch1 expression is involved in HL-60 cell growth inhibition induced by 4-hydroxynonenal, a product of lipid peroxidation
    • 2-s2.0-57349111927 10.2174/157340608786242098
    • Pizzimenti S., Barrera G., Calzavara E., Mirandola L., Toaldo C., Dianzani M. U., Comi P., Chiaramonte R., Down-regulation of Notch1 expression is involved in HL-60 cell growth inhibition induced by 4-hydroxynonenal, a product of lipid peroxidation. Medicinal Chemistry 2008 4 6 551 557 2-s2.0-57349111927 10.2174/157340608786242098
    • (2008) Medicinal Chemistry , vol.4 , Issue.6 , pp. 551-557
    • Pizzimenti, S.1    Barrera, G.2    Calzavara, E.3    Mirandola, L.4    Toaldo, C.5    Dianzani, M.U.6    Comi, P.7    Chiaramonte, R.8
  • 336
    • 3343002135 scopus 로고    scopus 로고
    • 4-Hydroxynonenal and regulation of cell cycle: Effects on the pRb/E2F pathway
    • DOI 10.1016/j.freeradbiomed.2004.05.023, PII S0891584904004277
    • Barrera G., Pizzimenti S., Dianzani M. U., 4-Hydroxynonenal and regulation of cell cycle: effects on the pRb/E2F pathway. Free Radical Biology and Medicine 2004 37 5 597 606 2-s2.0-3343002135 10.1016/j.freeradbiomed.2004. 05.023 (Pubitemid 38993508)
    • (2004) Free Radical Biology and Medicine , vol.37 , Issue.5 , pp. 597-606
    • Barrera, G.1    Pizzimenti, S.2    Dianzani, M.U.3
  • 337
    • 0029871935 scopus 로고    scopus 로고
    • Effect of 4-hydroxynonenal on cell cycle progression and expression of differentiation-associated antigens in HL-60 cells
    • DOI 10.1016/0891-5849(95)02049-7
    • Barrera G., Pizzimenti S., Muraca R., Barbiero G., Bonelli G., Baccino F. M., Fazio V. M., Dianzani M. U., Effect of 4-hydroxynonenal on cell cycle progression and expression of differentiation-associated antigens in HL-60 cells. Free Radical Biology and Medicine 1996 20 3 455 462 2-s2.0-0029871935 10.1016/0891-5849(95)02049-7 (Pubitemid 26077869)
    • (1996) Free Radical Biology and Medicine , vol.20 , Issue.3 , pp. 455-462
    • Barrera, G.1    Pizzimenti, S.2    Muraca, R.3    Barbiero, G.4    Bonelli, G.5    Baccino, F.M.6    Fazio, V.M.7    Dianzani, M.U.8
  • 338
    • 84880052663 scopus 로고    scopus 로고
    • 4-Hydroxynonenal induces G2/M phase cell cycle arrest by activation of the ataxia telangiectasia mutated and Rad3-related protein (ATR)/checkpoint kinase 1 (Chk1) signaling pathway
    • Chaudhary P., Sharma R., Sahu M., Vishwanatha J. K., Awasthi S., Awasthi Y. C., 4-Hydroxynonenal induces G2/M phase cell cycle arrest by activation of the ataxia telangiectasia mutated and Rad3-related protein (ATR)/checkpoint kinase 1 (Chk1) signaling pathway. Journal of Biological Chemistry 2013 288 28 20532 20546
    • (2013) Journal of Biological Chemistry , vol.288 , Issue.28 , pp. 20532-20546
    • Chaudhary, P.1    Sharma, R.2    Sahu, M.3    Vishwanatha, J.K.4    Awasthi, S.5    Awasthi, Y.C.6
  • 339
    • 84862780548 scopus 로고    scopus 로고
    • 4-hydroxy-2-nonenal mediates genotoxicity and bystander effects caused by enterococcus faecalis-infected macrophages
    • 2-s2.0-84857631398 10.1053/j.gastro.2011.11.020
    • Wang X., Yang Y., Moore D. R., Nimmo S. L., Lightfoot S. A., Huycke M. M., 4-hydroxy-2-nonenal mediates genotoxicity and bystander effects caused by enterococcus faecalis-infected macrophages. Gastroenterology 2012 142 3 543 551 2-s2.0-84857631398 10.1053/j.gastro.2011.11.020
    • (2012) Gastroenterology , vol.142 , Issue.3 , pp. 543-551
    • Wang, X.1    Yang, Y.2    Moore, D.R.3    Nimmo, S.L.4    Lightfoot, S.A.5    Huycke, M.M.6
  • 341
    • 34848876430 scopus 로고    scopus 로고
    • Deciphering the mechanism of HNE-induced apoptosis in cultured murine cortical neurons: Transcriptional responses and cellular pathways
    • DOI 10.1016/j.neuropharm.2007.07.016, PII S0028390807002444
    • Peng Z. F., Koh C. H. V., Li Q. T., Manikandan J., Melendez A. J., Tang S. Y., Halliwell B., Cheung N. S., Deciphering the mechanism of HNE-induced apoptosis in cultured murine cortical neurons: transcriptional responses and cellular pathways. Neuropharmacology 2007 53 5 687 698 2-s2.0-34848876430 10.1016/j.neuropharm.2007.07.016 (Pubitemid 47513174)
    • (2007) Neuropharmacology , vol.53 , Issue.5 , pp. 687-698
    • Peng, Z.F.1    Koh, C.H.V.2    Li, Q.T.3    Manikandan, J.4    Melendez, A.J.5    Tang, S.Y.6    Halliwell, B.7    Cheung, N.S.8
  • 342
    • 33644878774 scopus 로고    scopus 로고
    • Age-related differential growth rate and response to 4-hydroxynonenal in mouse aortic smooth muscle cells
    • 2-s2.0-33644878774
    • Lee T.-J., Lee J.-T., Moon S.-K., Kim C.-H., Park J.-W., Kwon T. K., Age-related differential growth rate and response to 4-hydroxynonenal in mouse aortic smooth muscle cells. International Journal of Molecular Medicine 2006 17 1 29 35 2-s2.0-33644878774
    • (2006) International Journal of Molecular Medicine , vol.17 , Issue.1 , pp. 29-35
    • Lee, T.-J.1    Lee, J.-T.2    Moon, S.-K.3    Kim, C.-H.4    Park, J.-W.5    Kwon, T.K.6
  • 343
    • 0035967824 scopus 로고    scopus 로고
    • Vascular smooth muscle cell activation and growth by 4-hydroxynonenal
    • DOI 10.1016/S0024-3205(01)01166-3, PII S0024320501011663
    • Kakishita H., Hattori Y., Vascular smooth muscle cell activation and growth by 4-hydroxynonenal. Life Sciences 2001 69 6 689 697 2-s2.0-0035967824 10.1016/S0024-3205(01)01166-3 (Pubitemid 32673627)
    • (2001) Life Sciences , vol.69 , Issue.6 , pp. 689-697
    • Kakishita, H.1    Hattori, Y.2
  • 344
    • 77953169082 scopus 로고    scopus 로고
    • Aldose reductase regulates vascular smooth muscle cell proliferation by modulating G1/S phase transition of cell cycle
    • 2-s2.0-77953169082 10.1210/en.2010-0160
    • Tammali R., Saxena A., Srivastava S. K., Ramana K. V., Aldose reductase regulates vascular smooth muscle cell proliferation by modulating G1/S phase transition of cell cycle. Endocrinology 2010 151 5 2140 2150 2-s2.0-77953169082 10.1210/en.2010-0160
    • (2010) Endocrinology , vol.151 , Issue.5 , pp. 2140-2150
    • Tammali, R.1    Saxena, A.2    Srivastava, S.K.3    Ramana, K.V.4
  • 345
    • 1542328799 scopus 로고    scopus 로고
    • Human neutrophil-derived elastase induces airway smooth muscle cell proliferation
    • DOI 10.1016/j.lfs.2003.07.059, PII S0024320504000475
    • Huang C.-D., Chen H.-H., Wang C.-H., Chou C.-L., Lin S.-M., Lin H.-C., Kuo H.-P., Human neutrophil-derived elastase induces airway smooth muscle cell proliferation. Life Sciences 2004 74 20 2479 2492 2-s2.0-1542328799 10.1016/j.lfs.2003.07.059 (Pubitemid 38317113)
    • (2004) Life Sciences , vol.74 , Issue.20 , pp. 2479-2492
    • Huang, C.-D.1    Chen, H.-H.2    Wang, C.-H.3    Chou, C.-L.4    Lin, S.-M.5    Lin, H.-C.6    Kuo, H.-P.7
  • 346
    • 79952275493 scopus 로고    scopus 로고
    • The "two-faced" effects of reactive oxygen species and the lipid peroxidation product 4-hydroxynonenal in the hallmarks of cancer
    • 2-s2.0-79952275493 10.3390/cancers2020338
    • Pizzimenti S., Toaldo C., Pettazzoni P., Dianzani M. U., Barrera G., The "two-faced" effects of reactive oxygen species and the lipid peroxidation product 4-hydroxynonenal in the hallmarks of cancer. Cancers 2010 2 2 338 363 2-s2.0-79952275493 10.3390/cancers2020338
    • (2010) Cancers , vol.2 , Issue.2 , pp. 338-363
    • Pizzimenti, S.1    Toaldo, C.2    Pettazzoni, P.3    Dianzani, M.U.4    Barrera, G.5
  • 347
    • 67650071137 scopus 로고    scopus 로고
    • Targeting cancer cells by ROS-mediated mechanisms: A radical therapeutic approach?
    • 2-s2.0-67650071137 10.1038/nrd2803
    • Trachootham D., Alexandre J., Huang P., Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Nature Reviews Drug Discovery 2009 8 7 579 591 2-s2.0-67650071137 10.1038/nrd2803
    • (2009) Nature Reviews Drug Discovery , vol.8 , Issue.7 , pp. 579-591
    • Trachootham, D.1    Alexandre, J.2    Huang, P.3
  • 348
    • 2942593991 scopus 로고    scopus 로고
    • ROS stress in cancer cells and therapeutic implications
    • DOI 10.1016/j.drup.2004.01.004
    • Pelicano H., Carney D., Huang P., ROS stress in cancer cells and therapeutic implications. Drug Resistance Updates 2004 7 2 97 110 2-s2.0-2942593991 10.1016/j.drup.2004.01.004 (Pubitemid 38736063)
    • (2004) Drug Resistance Updates , vol.7 , Issue.2 , pp. 97-110
    • Pelicano, H.1    Carney, D.2    Huang, P.3
  • 349
    • 1442360000 scopus 로고    scopus 로고
    • Intrinsic oxidative stress in cancer cells: A biochemical basis for therapeutic selectivity
    • DOI 10.1007/s00280-003-0726-5
    • Hileman E. O., Liu J., Albitar M., Keating M. J., Huang P., Intrinsic oxidative stress in cancer cells: a biochemical basis for therapeutic selectivity. Cancer Chemotherapy and Pharmacology 2004 53 3 209 219 2-s2.0-1442360000 10.1007/s00280-003-0726-5 (Pubitemid 38293718)
    • (2004) Cancer Chemotherapy and Pharmacology , vol.53 , Issue.3 , pp. 209-219
    • Hileman, E.O.1    Liu, J.2    Albitar, M.3    Keating, M.J.4    Huang, P.5
  • 351
    • 0035936797 scopus 로고    scopus 로고
    • The TNF and TNF receptor superfamilies: Integrating mammalian biology
    • DOI 10.1016/S0092-8674(01)00237-9
    • Locksley R. M., Killeen N., Lenardo M. J., The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell 2001 104 4 487 501 2-s2.0-0035936797 10.1016/S0092-8674(01)00237-9 (Pubitemid 32201945)
    • (2001) Cell , vol.104 , Issue.4 , pp. 487-501
    • Locksley, R.M.1    Killeen, N.2    Lenardo, M.J.3
  • 352
    • 34250308322 scopus 로고    scopus 로고
    • Apoptosis: A review of programmed cell death
    • DOI 10.1080/01926230701320337, PII 779478428
    • Elmore S., Apoptosis: a review of programmed cell death. Toxicologic Pathology 2007 35 4 495 516 (Pubitemid 46911891)
    • (2007) Toxicologic Pathology , vol.35 , Issue.4 , pp. 495-516
    • Elmore, S.1
  • 353
    • 79952825980 scopus 로고    scopus 로고
    • Redox regulation of the intrinsic pathway in neuronal apoptosis
    • 2-s2.0-79952825980 10.1089/ars.2010.3596
    • Franklin J. L., Redox regulation of the intrinsic pathway in neuronal apoptosis. Antioxidants and Redox Signaling 2011 14 8 1437 1448 2-s2.0-79952825980 10.1089/ars.2010.3596
    • (2011) Antioxidants and Redox Signaling , vol.14 , Issue.8 , pp. 1437-1448
    • Franklin, J.L.1
  • 354
    • 0242319666 scopus 로고    scopus 로고
    • Apoptosis - The p53 network
    • DOI 10.1242/jcs.00739
    • Haupt S., Berger M., Goldberg Z., Haupt Y., Apoptosis-the p53 network. Journal of Cell Science 2003 116 20 4077 4085 2-s2.0-0242319666 10.1242/jcs.00739 (Pubitemid 37337082)
    • (2003) Journal of Cell Science , vol.116 , Issue.20 , pp. 4077-4085
    • Haupt, S.1    Berger, M.2    Goldberg, Z.3    Haupt, Y.4
  • 355
    • 84859855659 scopus 로고    scopus 로고
    • 4-hydroxynonenal induces mitochondrial-mediated apoptosis and oxidative stress in SH-SY5Y human neuronal cells
    • 2-s2.0-84859855659 10.1111/j.1742-7843.2011.00834.x
    • Abarikwu S. O., Pant A. B., Farombi E. O., 4-hydroxynonenal induces mitochondrial-mediated apoptosis and oxidative stress in SH-SY5Y human neuronal cells. Basic and Clinical Pharmacology and Toxicology 2012 110 5 441 448 2-s2.0-84859855659 10.1111/j.1742-7843.2011.00834.x
    • (2012) Basic and Clinical Pharmacology and Toxicology , vol.110 , Issue.5 , pp. 441-448
    • Abarikwu, S.O.1    Pant, A.B.2    Farombi, E.O.3
  • 357
    • 37848999061 scopus 로고    scopus 로고
    • 4-hydroxynonenal self-limits Fas-mediated DISC-independent apoptosis by promoting export of Daxx from the nucleus to the cytosol and its binding to Fas
    • 2-s2.0-37848999061 10.1021/bi701559f
    • Sharma R., Sharma A., Dwivedi S., Zimniak P., Awasthi S., Awasthi Y. C., 4-hydroxynonenal self-limits Fas-mediated DISC-independent apoptosis by promoting export of Daxx from the nucleus to the cytosol and its binding to Fas. Biochemistry 2008 47 1 143 156 2-s2.0-37848999061 10.1021/bi701559f
    • (2008) Biochemistry , vol.47 , Issue.1 , pp. 143-156
    • Sharma, R.1    Sharma, A.2    Dwivedi, S.3    Zimniak, P.4    Awasthi, S.5    Awasthi, Y.C.6
  • 358
    • 54749095969 scopus 로고    scopus 로고
    • 4-hydroxynonenal induces apoptosis in human osteoarthritic chondrocytes: The protective role of glutathione-S-transferase
    • ARTICLE R107 2-s2.0-54749095969 10.1186/ar2503
    • Vaillancourt F., Fahmi H., Shi Q., Lavigne P., Ranger P., Fernandes J. C., Benderdour M., 4-hydroxynonenal induces apoptosis in human osteoarthritic chondrocytes: the protective role of glutathione-S-transferase. Arthritis Research and Therapy 2008 10 5, article R107 2-s2.0-54749095969 10.1186/ar2503
    • (2008) Arthritis Research and Therapy , vol.10 , Issue.5
    • Vaillancourt, F.1    Fahmi, H.2    Shi, Q.3    Lavigne, P.4    Ranger, P.5    Fernandes, J.C.6    Benderdour, M.7
  • 359
    • 44649173705 scopus 로고    scopus 로고
    • Self-regulatory role of 4-hydroxynonenal in signaling for stress-induced programmed cell death
    • 2-s2.0-44649173705 10.1016/j.freeradbiomed.2008.04.007
    • Awasthi Y. C., Sharma R., Sharma A., Yadav S., Singhal S. S., Chaudhary P., Awasthi S., Self-regulatory role of 4-hydroxynonenal in signaling for stress-induced programmed cell death. Free Radical Biology and Medicine 2008 45 2 111 118 2-s2.0-44649173705 10.1016/j.freeradbiomed.2008.04.007
    • (2008) Free Radical Biology and Medicine , vol.45 , Issue.2 , pp. 111-118
    • Awasthi, Y.C.1    Sharma, R.2    Sharma, A.3    Yadav, S.4    Singhal, S.S.5    Chaudhary, P.6    Awasthi, S.7
  • 360
    • 0036852609 scopus 로고    scopus 로고
    • Covalent modification of amino acid nucleophiles by the lipid peroxidation products 4-hydroxy-2-nonenal and 4-oxo-2-nonenal
    • DOI 10.1021/tx025590o
    • Doorn J. A., Petersen D. R., Covalent modification of amino acid nucleophiles by the lipid peroxidation products 4-hydroxy-2-nonenal and 4-oxo-2-nonenal. Chemical Research in Toxicology 2002 15 11 1445 1450 2-s2.0-0036852609 10.1021/tx025590o (Pubitemid 35364906)
    • (2002) Chemical Research in Toxicology , vol.15 , Issue.11 , pp. 1445-1450
    • Doorn, J.A.1    Petersen, D.R.2
  • 361
    • 33845390864 scopus 로고    scopus 로고
    • Protein adducts generated from products of lipid oxidation: Focus on HNE and ONE
    • DOI 10.1080/03602530600959508, PII X078211547323X23
    • Sayre L. M., Lin D., Yuan Q., Zhu X., Tang X., Protein adducts generated from products of lipid oxidation: focus on HNE and ONE. Drug Metabolism Reviews 2006 38 4 651 675 2-s2.0-33845390864 10.1080/03602530600959508 (Pubitemid 44903261)
    • (2006) Drug Metabolism Reviews , vol.38 , Issue.4 , pp. 651-675
    • Sayre, L.M.1    Lin, D.2    Yuan, Q.3    Zhu, X.4    Tang, X.5
  • 362
    • 84890507960 scopus 로고    scopus 로고
    • Modification and functional inhibition of regulator of G-protein signaling 4 (RGS4) by 4-Hydroxy-2-nonenal
    • Monroy C. A., Doorn J. A., Roman D. L., Modification and functional inhibition of regulator of G-protein signaling 4 (RGS4) by 4-Hydroxy-2-nonenal. Chemical Research in Toxicology 2013 26 12 1832 1839
    • (2013) Chemical Research in Toxicology , vol.26 , Issue.12 , pp. 1832-1839
    • Monroy, C.A.1    Doorn, J.A.2    Roman, D.L.3
  • 363
    • 45249124663 scopus 로고    scopus 로고
    • 4-Hydroxynonenal: A membrane lipid oxidation product of medicinal interest
    • DOI 10.1002/med.20117
    • Poli G., Schaur R. J., Siems W. G., Leonarduzzi G., 4-hydroxynonenal: a membrane lipid oxidation product of medicinal interest. Medicinal Research Reviews 2008 28 4 569 631 2-s2.0-45249124663 10.1002/med.20117 (Pubitemid 351842143)
    • (2008) Medicinal Research Reviews , vol.28 , Issue.4 , pp. 569-631
    • Poli, G.1    Schaur, R.J.2    Siems, W.G.3    Leonarduzzi, G.4
  • 364
    • 2942593925 scopus 로고    scopus 로고
    • 2-propanodeoxyguanine DNA adducts by human cell nuclear extracts
    • DOI 10.1021/bi049877r
    • 2-propanodeoxyguanine DNA adducts by human cell nuclear extracts. Biochemistry 2004 43 23 7514 7521 2-s2.0-2942593925 10.1021/bi049877r (Pubitemid 38745767)
    • (2004) Biochemistry , vol.43 , Issue.23 , pp. 7514-7521
    • Choudhury, S.1    Pan, J.2    Amin, S.3    Chung, F.-L.4    Roy, R.5
  • 365
    • 84888030895 scopus 로고    scopus 로고
    • Repair kinetics of acrolein- and (E)-4-hydroxy-2-nonenal-derived DNA adducts in human colon cell extracts
    • Choudhury S., Dyba M., Pan J., Roy R., Chung F. L., Repair kinetics of acrolein- and (E)-4-hydroxy-2-nonenal-derived DNA adducts in human colon cell extracts. Mutation Research 2013 751-752 15 23
    • (2013) Mutation Research , vol.751-752 , pp. 15-23
    • Choudhury, S.1    Dyba, M.2    Pan, J.3    Roy, R.4    Chung, F.L.5
  • 368
    • 33745847542 scopus 로고    scopus 로고
    • Increased etheno-DNA adducts in affected tissues of patients suffering from Crohn's disease, ulcerative colitis, and chronic pancreatitis
    • DOI 10.1089/ars.2006.8.1003
    • Nair J., Gansauge F., Beger H., Dolara P., Winde G., Bartsch H., Increased etheno-DNA adducts in affected tissues of patients suffering from Crohn's disease, ulcerative colitis, and chronic pancreatitis. Antioxidants and Redox Signaling 2006 8 5-6 1003 1010 2-s2.0-33745847542 10.1089/ars.2006.8.1003 (Pubitemid 44036517)
    • (2006) Antioxidants and Redox Signaling , vol.8 , Issue.5-6 , pp. 1003-1010
    • Nair, J.1    Gansauge, F.2    Beger, H.3    Dolara, P.4    Winde, G.5    Bartsch, H.6
  • 370
    • 0029841946 scopus 로고    scopus 로고
    • Effect of oxidative stress, produced by cumene hydroperoxide, on the various steps of protein synthesis. Modifications of elongation factor-2
    • DOI 10.1074/jbc.271.38.23105
    • Ayala A., Parrado J., Bougria M., Machado A., Effect of oxidative stress, produced by cumene hydroperoxide, on the various steps of protein synthesis. Modifications of elongation factor-2. Journal of Biological Chemistry 1996 271 38 23105 23110 2-s2.0-0029841946 10.1074/jbc.271.38.23105 (Pubitemid 26314744)
    • (1996) Journal of Biological Chemistry , vol.271 , Issue.38 , pp. 23105-23110
    • Ayala, A.1    Parrado, J.2    Bougria, M.3    Machado, A.4
  • 371
    • 0032901078 scopus 로고    scopus 로고
    • Effects of aging on the various steps of protein synthesis: Fragmentation of elongation factor 2
    • DOI 10.1016/S0891-5849(98)00202-0, PII S0891584998002020
    • Parrado J., Bougria M., Ayala A., Castaño A., Machado A., Effects of aging on the various steps of protein synthesis: fragmentation of elongation factor 2. Free Radical Biology and Medicine 1999 26 3-4 362 370 2-s2.0-0032901078 10.1016/S0891-5849(98)00202-0 (Pubitemid 29013468)
    • (1999) Free Radical Biology and Medicine , vol.26 , Issue.3-4 , pp. 362-370
    • Parrado, J.1    Bougria, M.2    Ayala, A.3    Castano, A.4    Machado, A.5
  • 372
    • 0033028705 scopus 로고    scopus 로고
    • Induced mono-(ADP)-ribosylation of rat liver cytosolic proteins by lipid peroxidant agents
    • DOI 10.1016/S0891-5849(98)00316-5, PII S0891584998003165
    • Parrado J., Bougria M., Ayala A., MacHado A., Induced mono-(ADP)-ribosylation of rat liver cytosolic proteins by lipid peroxidant agents. Free Radical Biology and Medicine 1999 26 9-10 1079 1084 2-s2.0-0033028705 10.1016/S0891-5849(98)00316-5 (Pubitemid 29262691)
    • (1999) Free Radical Biology and Medicine , vol.26 , Issue.9-10 , pp. 1079-1084
    • Parrado, J.1    Bougria, M.2    Ayala, A.3    MacHado, A.4
  • 373
    • 0344827189 scopus 로고    scopus 로고
    • "In vitro" effect of cumene hydroperoxide on hepatic elongation factor-2 and its protection by melatonin
    • DOI 10.1016/j.bbagen.2003.10.006
    • Parrado J., Absi E. H., Machado A., Ayala A., 'In vitro' effect of cumene hydroperoxide on hepatic elongation factor-2 and its protection by melatonin. Biochimica et Biophysica Acta: General Subjects 2003 1624 1-3 139 144 2-s2.0-0344827189 10.1016/j.bbagen.2003.10.006 (Pubitemid 37464951)
    • (2003) Biochimica et Biophysica Acta - General Subjects , vol.1624 , Issue.1-3 , pp. 139-144
    • Parrado, J.1    Absi, E.H.2    Machado, A.3    Ayala, A.4
  • 374
    • 33644903770 scopus 로고    scopus 로고
    • In vitro effect of lipid peroxidation metabolites on elongation factor-2
    • 2-s2.0-33644903770 10.1016/j.bbagen.2005.12.019
    • Argüelles S., Machado A., Ayala A., 'In vitro' effect of lipid peroxidation metabolites on elongation factor-2. Biochimica et Biophysica Acta: General Subjects 2006 1760 3 445 452 2-s2.0-33644903770 10.1016/j.bbagen.2005. 12.019
    • (2006) Biochimica et Biophysica Acta: General Subjects , vol.1760 , Issue.3 , pp. 445-452
    • Argüelles, S.1    Machado, A.2    Ayala, A.3
  • 375
    • 79251595904 scopus 로고    scopus 로고
    • Effect of aging and oxidative stress on elongation factor-2 in hypothalamus and hypophysis
    • 2-s2.0-79251595904 10.1016/j.mad.2010.12.002
    • Arguelles S., Cano M., Machado A., Ayala A., Effect of aging and oxidative stress on elongation factor-2 in hypothalamus and hypophysis. Mechanisms of Ageing and Development 2011 132 1-2 55 64 2-s2.0-79251595904 10.1016/j.mad.2010.12.002
    • (2011) Mechanisms of Ageing and Development , vol.132 , Issue.1-2 , pp. 55-64
    • Arguelles, S.1    Cano, M.2    Machado, A.3    Ayala, A.4
  • 376
    • 84863593560 scopus 로고    scopus 로고
    • In vitro and in vivo protection by melatonin against the decline of elongation factor-2 caused by lipid peroxidation: Preservation of protein synthesis
    • Argüelles S., Muñoz M. F., Cano M., Machado A., Ayala A., In vitro and in vivo protection by melatonin against the decline of elongation factor-2 caused by lipid peroxidation: preservation of protein synthesis. Journal of Pineal Research 2012 53 1 1 10
    • (2012) Journal of Pineal Research , vol.53 , Issue.1 , pp. 1-10
    • Argüelles, S.1    Muñoz, M.F.2    Cano, M.3    Machado, A.4    Ayala, A.5
  • 377
    • 34548062369 scopus 로고    scopus 로고
    • In vitro protective effect of a hydrophilic vitamin E analogue on the decrease in levels of elongation factor 2 in conditions of oxidative stress
    • DOI 10.1159/000102948
    • Argüelles S., Machado A., Ayala A., 'In vitro' protective effect of a hydrophilic vitamin E analogue on the decrease in levels of elongation factor 2 in conditions of oxidative stress. Gerontology 2007 53 5 282 288 2-s2.0-34548062369 10.1159/000102948 (Pubitemid 47294452)
    • (2007) Gerontology , vol.53 , Issue.5 , pp. 282-288
    • Arguelles, S.1    Machado, A.2    Ayala, A.3
  • 378
    • 77955194558 scopus 로고    scopus 로고
    • Comparative study of the in Vitro protective effects of several antioxidants on elongation factor 2 under oxidative stress conditions
    • 2-s2.0-77955194558 10.1271/bbb.100054
    • Arguelles S., Cano M., Machado A., Ayala A., Comparative study of the In Vitro protective effects of several antioxidants on elongation factor 2 under oxidative stress conditions. Bioscience, Biotechnology and Biochemistry 2010 74 7 1373 1379 2-s2.0-77955194558 10.1271/bbb.100054
    • (2010) Bioscience, Biotechnology and Biochemistry , vol.74 , Issue.7 , pp. 1373-1379
    • Arguelles, S.1    Cano, M.2    Machado, A.3    Ayala, A.4
  • 379
    • 84877039778 scopus 로고    scopus 로고
    • Molecular control of the amount, subcellular location, and activity state of translation elongation factor 2 in neurons experiencing stress
    • Argüelles S., Camandola S., Hutchison E. R., Cutler R. G., Ayala A., Mattson M. P., Molecular control of the amount, subcellular location, and activity state of translation elongation factor 2 in neurons experiencing stress. Free Radical Biology and Medicine 2013 61 61 71
    • (2013) Free Radical Biology and Medicine , vol.61 , pp. 61-71
    • Argüelles, S.1    Camandola, S.2    Hutchison, E.R.3    Cutler, R.G.4    Ayala, A.5    Mattson, M.P.6
  • 380
    • 84888125439 scopus 로고    scopus 로고
    • Elongation factor 2 diphthamide is critical for translation of two IRES-dependent protein targets, XIAP and FGF2, under oxidative stress conditions
    • Argüelles S., Camandola S., Cutler R. G., Ayala A., Mattson M. P., Elongation factor 2 diphthamide is critical for translation of two IRES-dependent protein targets, XIAP and FGF2, under oxidative stress conditions. Free Radical Biology and Medicine 2013 67 131 138
    • (2013) Free Radical Biology and Medicine , vol.67 , pp. 131-138
    • Argüelles, S.1    Camandola, S.2    Cutler, R.G.3    Ayala, A.4    Mattson, M.P.5
  • 381
    • 84861494710 scopus 로고    scopus 로고
    • A red palm oil diet can reduce the effects of oxidative stress on rat spermatozoa
    • supplement 1
    • Aboua Y. G., Brooks N., Mahfouz R. Z., Agarwal A., du Plessis S. S., A red palm oil diet can reduce the effects of oxidative stress on rat spermatozoa. Andrologia 2012 44 supplement 1 32 40
    • (2012) Andrologia , vol.44 , pp. 32-40
    • Aboua, Y.G.1    Brooks, N.2    Mahfouz, R.Z.3    Agarwal, A.4    Du Plessis, S.S.5
  • 382
    • 33846018570 scopus 로고    scopus 로고
    • Induction of oxidative stress by organic hydroperoxides in testis and epididymal sperm of rats in vivo
    • DOI 10.2164/jandrol.106.000265
    • Kumar T. R., Muralidhara M., Induction of oxidative stress by organic hydroperoxides in testis and epididymal sperm of rats in vivo. Journal of Andrology 2007 28 1 77 85 2-s2.0-33846018570 10.2164/jandrol.106.000265 (Pubitemid 46048081)
    • (2007) Journal of Andrology , vol.28 , Issue.1 , pp. 77-85
    • Kumar, T.R.1    Muralidhara2
  • 383
    • 14644439227 scopus 로고    scopus 로고
    • The biosynthesis of ascorbate protects isolated rat hepatocytes from cumene hydroperoxide-mediated oxidative stress
    • DOI 10.1016/j.freeradbiomed.2004.12.006
    • Chan T. S., Shangari N., Wilson J. X., Chan H., Butterworth R. F., O'Brien P. J., The biosynthesis of ascorbate protects isolated rat hepatocytes from cumene hydroperoxide-mediated oxidative stress. Free Radical Biology and Medicine 2005 38 7 867 873 2-s2.0-14644439227 10.1016/j.freeradbiomed.2004.12. 006 (Pubitemid 40321078)
    • (2005) Free Radical Biology and Medicine , vol.38 , Issue.7 , pp. 867-873
    • Chan, T.S.1    Shangari, N.2    Wilson, J.X.3    Chan, H.4    Butterworth, R.F.5    O'Brien, P.J.6
  • 384
    • 0036851874 scopus 로고    scopus 로고
    • Antioxidant balance and free radical generation in vitamin E-deficient mice after dermal exposure to cumene hydroperoxide
    • DOI 10.1021/tx0200313
    • Shvedova A. A., Kisin E. R., Murray A. R., Kommineni C., Castranova V., Mason R. P., Kadiiska M. B., Gunther M. R., Antioxidant balance and free radical generation in vitamin E-deficient mice after dermal exposure to cumene hydroperoxide. Chemical Research in Toxicology 2002 15 11 1451 1459 2-s2.0-0036851874 10.1021/tx0200313 (Pubitemid 35364907)
    • (2002) Chemical Research in Toxicology , vol.15 , Issue.11 , pp. 1451-1459
    • Shvedova, A.A.1    Kisin, E.R.2    Murray, A.R.3    Kommineni, C.4    Castranova, V.5    Mason, R.P.6    Kadiiska, M.B.7    Gunther, M.R.8
  • 385
    • 0034072323 scopus 로고    scopus 로고
    • Myrica nagi attenuates cumene hydroperoxide-induced cutaneous oxidative stress and toxicity in Swiss albino mice
    • Alam A., Iqbal M., Saleem M., Ahmed S.-U., Sultana S., Myrica nagi attenuates cumene hydroperoxide-induced cutaneous oxidative stress and toxicity in Swiss albino mice. Pharmacology and Toxicology 2000 86 5 209 214 2-s2.0-0034072323 (Pubitemid 30333647)
    • (2000) Pharmacology and Toxicology , vol.86 , Issue.5 , pp. 209-214
    • Alam, A.1    Iqbal, M.2    Saleem, M.3    Ahmed, S.-U.4    Sultana, S.5
  • 387
    • 84871742397 scopus 로고    scopus 로고
    • Protective effect of 70% ethanolic extract of Lindera obtusiloba Blume on tert-butyl hydroperoxide-induced oxidative hepatotoxicity in rats
    • Hong C. O., Rhee C. H., Won N. H., Choi H. D., Lee K. W., Protective effect of 70% ethanolic extract of Lindera obtusiloba Blume on tert-butyl hydroperoxide-induced oxidative hepatotoxicity in rats. Food and Chemical Toxicology 2013 53 214 220
    • (2013) Food and Chemical Toxicology , vol.53 , pp. 214-220
    • Hong, C.O.1    Rhee, C.H.2    Won, N.H.3    Choi, H.D.4    Lee, K.W.5
  • 389
    • 34547125977 scopus 로고    scopus 로고
    • Protective effect of aqueous extract of Perilla frutescens on tert-butyl hydroperoxide-induced oxidative hepatotoxicity in rats
    • DOI 10.1016/j.fct.2007.03.009, PII S0278691507001093
    • Kim M.-K., Lee H.-S., Kim E.-J., Won N.-H., Chi Y.-M., Kim B.-C., Lee K.-W., Protective effect of aqueous extract of Perilla frutescens on tert-butyl hydroperoxide-induced oxidative hepatotoxicity in rats. Food and Chemical Toxicology 2007 45 9 1738 1744 2-s2.0-34547125977 10.1016/j.fct.2007.03.009 (Pubitemid 47102511)
    • (2007) Food and Chemical Toxicology , vol.45 , Issue.9 , pp. 1738-1744
    • Kim, M.-K.1    Lee, H.-S.2    Kim, E.-J.3    Won, N.-H.4    Chi, Y.-M.5    Kim, B.-C.6    Lee, K.-W.7
  • 390
    • 33748435753 scopus 로고    scopus 로고
    • Tertiary-butyl hydroperoxide induced oxidative stress and male reproductive activity in mice: Role of transcription factor NF-κB and testicular antioxidant enzymes
    • DOI 10.1016/j.reprotox.2006.03.017, PII S0890623806000955
    • Kaur P., Kaur G., Bansal M. P., Tertiary-butyl hydroperoxide induced oxidative stress and male reproductive activity in mice: role of transcription factor NF- B and testicular antioxidant enzymes. Reproductive Toxicology 2006 22 3 479 484 2-s2.0-33748435753 10.1016/j.reprotox.2006.03.017 (Pubitemid 44344457)
    • (2006) Reproductive Toxicology , vol.22 , Issue.3 , pp. 479-484
    • Kaur, P.1    Kaur, G.2    Bansal, M.P.3
  • 391
    • 0036230951 scopus 로고    scopus 로고
    • In vivo protective effect of protocatechuic acid on tert-butyl hydroperoxide-induced rat hepatotoxicity
    • Liu C. L., Wang J. M., Chu C. Y., Cheng M. T., Tseng T. H., In vivo protective effect of protocatechuic acid on tert-butyl hydroperoxide-induced rat hepatotoxicity. Food and Chemical Toxicology 2002 40 5 635 641
    • (2002) Food and Chemical Toxicology , vol.40 , Issue.5 , pp. 635-641
    • Liu, C.L.1    Wang, J.M.2    Chu, C.Y.3    Cheng, M.T.4    Tseng, T.H.5
  • 392
    • 0033789365 scopus 로고    scopus 로고
    • In vivo metabolism of tert-Butyl hydroperoxide to methyl radicals. EPR spin-trapping and DNA methylation studies
    • 2-s2.0-0033789365 10.1021/tx000130l
    • Hix S., Kadiiska M. B., Mason R. P., Augusto O., In vivo metabolism of tert-Butyl hydroperoxide to methyl radicals. EPR spin-trapping and DNA methylation studies. Chemical Research in Toxicology 2000 13 10 1056 1064 2-s2.0-0033789365 10.1021/tx000130l
    • (2000) Chemical Research in Toxicology , vol.13 , Issue.10 , pp. 1056-1064
    • Hix, S.1    Kadiiska, M.B.2    Mason, R.P.3    Augusto, O.4
  • 393
    • 84890245314 scopus 로고    scopus 로고
    • Hepatoprotective properties of sesamin against CCl4 induced oxidative stress-mediated apoptosis in mice via JNK pathway
    • Ma J. Q., Ding J., Zhang L., Liu C. M., Hepatoprotective properties of sesamin against CCl4 induced oxidative stress-mediated apoptosis in mice via JNK pathway. Food and Chemical Toxicology 2013 64 41 48
    • (2013) Food and Chemical Toxicology , vol.64 , pp. 41-48
    • Ma, J.Q.1    Ding, J.2    Zhang, L.3    Liu, C.M.4
  • 394
    • 84881045937 scopus 로고    scopus 로고
    • Effects of yam peel extract against carbon tetrachloride-induced hepatotoxicity in rats
    • Yeh Y. H., Hsieh Y. L., Lee Y. T., Effects of yam peel extract against carbon tetrachloride-induced hepatotoxicity in rats. Journal of Agricultural and Food Chemistry 2013 61 30 7387 7396
    • (2013) Journal of Agricultural and Food Chemistry , vol.61 , Issue.30 , pp. 7387-7396
    • Yeh, Y.H.1    Hsieh, Y.L.2    Lee, Y.T.3
  • 396
    • 79957852763 scopus 로고    scopus 로고
    • Protective effects of hyperoside against carbon tetrachloride-induced liver damage in mice
    • 2-s2.0-79957852763 10.1021/np200001x
    • Choi J.-H., Kim D.-W., Yun N., Choi J.-S., Islam M. N., Kim Y.-S., Lee S.-M., Protective effects of hyperoside against carbon tetrachloride-induced liver damage in mice. Journal of Natural Products 2011 74 5 1055 1060 2-s2.0-79957852763 10.1021/np200001x
    • (2011) Journal of Natural Products , vol.74 , Issue.5 , pp. 1055-1060
    • Choi, J.-H.1    Kim, D.-W.2    Yun, N.3    Choi, J.-S.4    Islam, M.N.5    Kim, Y.-S.6    Lee, S.-M.7
  • 398
    • 23044488162 scopus 로고    scopus 로고
    • Melatonin ameliorates carbon tetrachloride-induced hepatic fibrogenesis in rats via inhibition of oxidative stress
    • DOI 10.1016/j.lfs.2005.04.013, PII S0024320505004078
    • Wang H., Wei W., Wang N.-P., Gui S.-Y., Wu L., Sun W.-Y., Xu S.-Y., Melatonin ameliorates carbon tetrachloride-induced hepatic fibrogenesis in rats via inhibition of oxidative stress. Life Sciences 2005 77 15 1902 1915 2-s2.0-23044488162 10.1016/j.lfs.2005.04.013 (Pubitemid 41058993)
    • (2005) Life Sciences , vol.77 , Issue.15 , pp. 1902-1915
    • Wang, H.1    Wei, W.2    Wang, N.-P.3    Gui, S.-Y.4    Wu, L.5    Sun, W.-Y.6    Xu, S.-Y.7
  • 401
    • 67649414190 scopus 로고    scopus 로고
    • Impact of co administration of selenium and quinolinic acid in the rat's brain
    • 2-s2.0-67649414190 10.1016/j.brainres.2009.05.036
    • Sreekala S., Indira M., Impact of co administration of selenium and quinolinic acid in the rat's brain. Brain Research 2009 1281 101 107 2-s2.0-67649414190 10.1016/j.brainres.2009.05.036
    • (2009) Brain Research , vol.1281 , pp. 101-107
    • Sreekala, S.1    Indira, M.2
  • 402
    • 26944485506 scopus 로고    scopus 로고
    • Peripheral benzodiazepine receptor ligand PK11195 reduces microglial activation and neuronal death in quinolinic acid-injected rat striatum
    • DOI 10.1016/j.nbd.2005.04.010, PII S0969996105001208
    • Ryu J. K., Choi H. B., McLarnon J. G., Peripheral benzodiazepine receptor ligand PK11195 reduces microglial activation and neuronal death in quinolinic acid-injected rat striatum. Neurobiology of Disease 2005 20 2 550 561 2-s2.0-26944485506 10.1016/j.nbd.2005.04.010 (Pubitemid 41476281)
    • (2005) Neurobiology of Disease , vol.20 , Issue.2 , pp. 550-561
    • Ryu, J.K.1    Choi, H.B.2    McLarnon, J.G.3
  • 403
    • 84984569420 scopus 로고    scopus 로고
    • Ebselen blocks the quinolinic acid-induced production of thiobarbituric acid reactive species but does not prevent the behavioral alterations produced by intra-striatal quinolinic acid administration in the rat
    • DOI 10.1016/S0304-3940(01)02504-6, PII S0304394001025046
    • Rossato J. I., Zeni G., Mello C. F., Rubin M. A., Rocha J. B. T., Ebselen blocks the quinolinic acid-induced production of thiobarbituric acid reactive species but does not prevent the behavioral alterations produced by intra-striatal quinolinic acid administration in the rat. Neuroscience Letters 2002 318 3 137 140 2-s2.0-0036468684 10.1016/S0304-3940(01)02504-6 (Pubitemid 34136028)
    • (2002) Neuroscience Letters , vol.318 , Issue.3 , pp. 137-140
    • Rossato, J.I.1    Zeni, G.2    Mello, C.F.3    Rubin, M.A.4    Rocha, J.B.T.5
  • 406
    • 79954642381 scopus 로고    scopus 로고
    • Advances in metal-induced oxidative stress and human disease
    • 2-s2.0-79954642381 10.1016/j.tox.2011.03.001
    • Jomova K., Valko M., Advances in metal-induced oxidative stress and human disease. Toxicology 2011 283 2-3 65 87 2-s2.0-79954642381 10.1016/j.tox.2011. 03.001
    • (2011) Toxicology , vol.283 , Issue.2-3 , pp. 65-87
    • Jomova, K.1    Valko, M.2
  • 408
    • 84861590749 scopus 로고    scopus 로고
    • N-acetylcysteine attenuates copper overload-induced oxidative injury in brain of rat
    • 2-s2.0-84855581202 10.1007/s12011-012-9320-1
    • Özcelik D., Uzun H., Naziroglu M., N-acetylcysteine attenuates copper overload-induced oxidative injury in brain of rat. Biological Trace Element Research 2012 147 1-3 292 298 2-s2.0-84855581202 10.1007/s12011-012- 9320-1
    • (2012) Biological Trace Element Research , vol.147 , Issue.1-3 , pp. 292-298
    • Özcelik, D.1    Uzun, H.2    Naziroglu, M.3
  • 410
    • 35348978352 scopus 로고    scopus 로고
    • The effects of the wood preservative copper dimethyldithiocarbamate in the hippocampus of maternal and newborn Long-Evans rats
    • DOI 10.1016/j.toxlet.2007.08.017, PII S0378427407009459
    • Scharf B., Trombetta L. D., The effects of the wood preservative copper dimethyldithiocarbamate in the hippocampus of maternal and newborn Long-Evans rats. Toxicology Letters 2007 174 1-3 117 124 2-s2.0-35348978352 10.1016/j.toxlet.2007.08.017 (Pubitemid 47599864)
    • (2007) Toxicology Letters , vol.174 , Issue.1-3 , pp. 117-124
    • Scharf, B.1    Trombetta, L.D.2
  • 411
    • 70249112934 scopus 로고    scopus 로고
    • Pycnogenol prevents potassium dichromate (K2Cr2O7)-induced oxidative damage and nephrotoxicity in rats
    • 2-s2.0-70249112934 10.1016/j.cbi.2009.08.001
    • Parveen K., Khan M. R., Siddiqui W. A., Pycnogenol prevents potassium dichromate (K2Cr2O7)-induced oxidative damage and nephrotoxicity in rats. Chemico-Biological Interactions 2009 181 3 343 350 2-s2.0-70249112934 10.1016/j.cbi.2009.08.001
    • (2009) Chemico-Biological Interactions , vol.181 , Issue.3 , pp. 343-350
    • Parveen, K.1    Khan, M.R.2    Siddiqui, W.A.3
  • 413
    • 84888200505 scopus 로고    scopus 로고
    • The effects of flaxseed oil on cadmium-induced oxidative stress in rats
    • Karaca S., Eraslan G., The effects of flaxseed oil on cadmium-induced oxidative stress in rats. Biological Trace Element Research 2013 155 3 423 430
    • (2013) Biological Trace Element Research , vol.155 , Issue.3 , pp. 423-430
    • Karaca, S.1    Eraslan, G.2
  • 414
    • 84882967906 scopus 로고    scopus 로고
    • The protective effect of grape seed procyanidin extract against cadmium-induced renal oxidative damage in mice
    • Chen Q., Zhang R., Li W., Niu Y., Guo H., Liu X., Hou Y., Zhao L., The protective effect of grape seed procyanidin extract against cadmium-induced renal oxidative damage in mice. Environmental Toxicology and Pharmacology 2013 36 3 759 768
    • (2013) Environmental Toxicology and Pharmacology , vol.36 , Issue.3 , pp. 759-768
    • Chen, Q.1    Zhang, R.2    Li, W.3    Niu, Y.4    Guo, H.5    Liu, X.6    Hou, Y.7    Zhao, L.8
  • 415
    • 80054851875 scopus 로고    scopus 로고
    • Beneficial effect of hesperetin on cadmium induced oxidative stress in rats: An in vivo and in vitro study
    • 2-s2.0-80054851875
    • Leelavinothan P., Kalist S., Beneficial effect of hesperetin on cadmium induced oxidative stress in rats: an in vivo and in vitro study. European Review for Medical and Pharmacological Sciences 2011 15 9 992 1002 2-s2.0-80054851875
    • (2011) European Review for Medical and Pharmacological Sciences , vol.15 , Issue.9 , pp. 992-1002
    • Leelavinothan, P.1    Kalist, S.2
  • 416
    • 77949570689 scopus 로고    scopus 로고
    • Cadmium-induced lipid peroxidation and changes in antioxidant defense system in the rat testes: Protective role of coenzyme Q(10) and vitamin e
    • Ognjanović B. I., Marković S. D., Ethordević N. Z., Trbojević I. S., Stajn A. S., Saicić Z. S., Cadmium-induced lipid peroxidation and changes in antioxidant defense system in the rat testes: protective role of coenzyme Q(10) and vitamin E. Reproductive Toxicology 2010 29 2 191 197
    • (2010) Reproductive Toxicology , vol.29 , Issue.2 , pp. 191-197
    • Ognjanović, B.I.1    Marković, S.D.2    Ethordević, N.Z.3    Trbojević, I.S.4    Stajn, A.S.5    Saicić, Z.S.6
  • 417
    • 79959955771 scopus 로고    scopus 로고
    • Beneficial role of naringin, a flavanoid on nickel induced nephrotoxicity in rats
    • 2-s2.0-79959955771 10.1016/j.cbi.2011.05.003
    • Amudha K., Pari L., Beneficial role of naringin, a flavanoid on nickel induced nephrotoxicity in rats. Chemico-Biological Interactions 2011 193 1 57 64 2-s2.0-79959955771 10.1016/j.cbi.2011.05.003
    • (2011) Chemico-Biological Interactions , vol.193 , Issue.1 , pp. 57-64
    • Amudha, K.1    Pari, L.2
  • 418
    • 78649737499 scopus 로고    scopus 로고
    • Hepatoprotective role of naringin on nickel-induced toxicity in male Wistar rats
    • 2-s2.0-78649737499 10.1016/j.ejphar.2010.09.068
    • Pari L., Amudha K., Hepatoprotective role of naringin on nickel-induced toxicity in male Wistar rats. European Journal of Pharmacology 2011 650 1 364 370 2-s2.0-78649737499 10.1016/j.ejphar.2010.09.068
    • (2011) European Journal of Pharmacology , vol.650 , Issue.1 , pp. 364-370
    • Pari, L.1    Amudha, K.2
  • 419
    • 84878718295 scopus 로고    scopus 로고
    • Magnesium can protect against vanadium-induced lipid peroxidation in the hepatic tissue
    • 802734 10.1155/2013/802734
    • Scibior A., Gołȩbiowska D., Niedźwiecka I., Magnesium can protect against vanadium-induced lipid peroxidation in the hepatic tissue. Oxidative Medicine and Cellular Longevity 2013 2013 11 802734 10.1155/2013/802734
    • (2013) Oxidative Medicine and Cellular Longevity , vol.2013 , pp. 11
    • Scibior, A.1    Gołȩbiowska, D.2    Niedźwiecka, I.3
  • 420
    • 77955359527 scopus 로고    scopus 로고
    • Lipid peroxidation in the kidney of rats treated with v and/or Mg in drinking water
    • 2-s2.0-77955359527 10.1002/jat.1520
    • Ścibior A., Zaporowska H., Niedźwiecka I., Lipid peroxidation in the kidney of rats treated with V and/or Mg in drinking water. Journal of Applied Toxicology 2010 30 5 487 496 2-s2.0-77955359527 10.1002/jat.1520
    • (2010) Journal of Applied Toxicology , vol.30 , Issue.5 , pp. 487-496
    • Ścibior, A.1    Zaporowska, H.2    Niedźwiecka, I.3
  • 421
    • 30944434392 scopus 로고    scopus 로고
    • Combined effect of vanadium(V) and chromium(III) on lipid peroxidation in liver and kidney of rats
    • DOI 10.1016/j.cbi.2005.11.008, PII S0009279705003972
    • Ścibior A., Zaporowska H., Ostrowski J., Banach A., Combined effect of vanadium(V) and chromium(III) on lipid peroxidation in liver and kidney of rats. Chemico-Biological Interactions 2006 159 3 213 222 2-s2.0-30944434392 10.1016/j.cbi.2005.11.008 (Pubitemid 43112432)
    • (2006) Chemico-Biological Interactions , vol.159 , Issue.3 , pp. 213-222
    • Scibior, A.1    Zaporowska, H.2    Ostrowski, J.3    Banach, A.4
  • 422
    • 84984563862 scopus 로고    scopus 로고
    • Protective effect of Melissa officinalis aqueous extract against Mn-induced oxidative stress in chronically exposed mice
    • 2-s2.0-84155163943 10.1016/j.brainresbull.2011.10.003
    • Martins E. N., Pessano N. T. C., Leal L., Roos D. H., Folmer V., Puntel G. O., Rocha J. B. T., Aschner M., Ávila D. S., Puntel R. L., Protective effect of Melissa officinalis aqueous extract against Mn-induced oxidative stress in chronically exposed mice. Brain Research Bulletin 2012 87 1 74 79 2-s2.0-84155163943 10.1016/j.brainresbull.2011.10.003
    • (2012) Brain Research Bulletin , vol.87 , Issue.1 , pp. 74-79
    • Martins, E.N.1    Pessano, N.T.C.2    Leal, L.3    Roos, D.H.4    Folmer, V.5    Puntel, G.O.6    Rocha, J.B.T.7    Aschner, M.8    Ávila, D.S.9    Puntel, R.L.10
  • 423
    • 78349312428 scopus 로고    scopus 로고
    • Silymarin, a natural antioxidant, protects cerebral cortex against manganese-induced neurotoxicity in adult rats
    • 2-s2.0-78349312428 10.1007/s10534-010-9345-x
    • Chtourou Y., Fetoui H., Sefi M., Trabelsi K., Barkallah M., Boudawara T., Kallel H., Zeghal N., Silymarin, a natural antioxidant, protects cerebral cortex against manganese-induced neurotoxicity in adult rats. BioMetals 2010 23 6 985 996 2-s2.0-78349312428 10.1007/s10534-010-9345-x
    • (2010) BioMetals , vol.23 , Issue.6 , pp. 985-996
    • Chtourou, Y.1    Fetoui, H.2    Sefi, M.3    Trabelsi, K.4    Barkallah, M.5    Boudawara, T.6    Kallel, H.7    Zeghal, N.8
  • 424
    • 33745698058 scopus 로고    scopus 로고
    • Effects of acute manganese chloride exposure on lipid peroxidation and alteration of trace metals in rat brain
    • Chen M. T., Cheng G. W., Lin C. C., Chen B. H., Huang Y. L., Effects of acute manganese chloride exposure on lipid peroxidation and alteration of trace metals in rat brain. Biological Trace Element Research 2006 110 2 163 178
    • (2006) Biological Trace Element Research , vol.110 , Issue.2 , pp. 163-178
    • Chen, M.T.1    Cheng, G.W.2    Lin, C.C.3    Chen, B.H.4    Huang, Y.L.5
  • 427
    • 84876077830 scopus 로고    scopus 로고
    • Dietary iron concentration may influence aging process by altering oxidative stress in tissues of adult rats
    • e61058
    • Arruda L. F., Arruda S. F., Campos N. A., de Valencia F. F., de Siqueira E. M., Dietary iron concentration may influence aging process by altering oxidative stress in tissues of adult rats. PloS ONE 2013 8 4 e61058
    • (2013) PloS ONE , vol.8 , Issue.4
    • Arruda, L.F.1    Arruda, S.F.2    Campos, N.A.3    De Valencia, F.F.4    De Siqueira, E.M.5
  • 428
    • 78349268336 scopus 로고    scopus 로고
    • Anti-inflammatory effects of pioglitazone on iron-induced oxidative injury in the nigrostriatal dopaminergic system
    • 2-s2.0-78349268336 10.1111/j.1365-2990.2010.01107.x
    • Yu H. C., Feng S. F., Chao P. L., Lin A. M. Y., Anti-inflammatory effects of pioglitazone on iron-induced oxidative injury in the nigrostriatal dopaminergic system. Neuropathology and Applied Neurobiology 2010 36 7 612 622 2-s2.0-78349268336 10.1111/j.1365-2990.2010.01107.x
    • (2010) Neuropathology and Applied Neurobiology , vol.36 , Issue.7 , pp. 612-622
    • Yu, H.C.1    Feng, S.F.2    Chao, P.L.3    Lin, A.M.Y.4
  • 431
    • 33644839704 scopus 로고    scopus 로고
    • Melatonin and 6-hydroxymelatonin protect against iron-induced neurotoxicity
    • 2-s2.0-33644839704 10.1111/j.1471-4159.2005.03532.x
    • Maharaj D. S., Maharaj H., Daya S., Glass B. D., Melatonin and 6-hydroxymelatonin protect against iron-induced neurotoxicity. Journal of Neurochemistry 2006 96 1 78 81 2-s2.0-33644839704 10.1111/j.1471-4159.2005. 03532.x
    • (2006) Journal of Neurochemistry , vol.96 , Issue.1 , pp. 78-81
    • Maharaj, D.S.1    Maharaj, H.2    Daya, S.3    Glass, B.D.4
  • 432
    • 84863543125 scopus 로고    scopus 로고
    • Hepatic reduction of carbamoyl-PROXYL in ferric nitrilotriacetate induced iron overloaded mice: An in vivo ESR study
    • Morales N. P., Yamaguchi Y., Murakami K., Kosem N., Utsumi H., Hepatic reduction of carbamoyl-PROXYL in ferric nitrilotriacetate induced iron overloaded mice: an in vivo ESR study. Biological and Pharmaceutical Bulletin 2012 35 7 1035 1040
    • (2012) Biological and Pharmaceutical Bulletin , vol.35 , Issue.7 , pp. 1035-1040
    • Morales, N.P.1    Yamaguchi, Y.2    Murakami, K.3    Kosem, N.4    Utsumi, H.5
  • 433
    • 18844408406 scopus 로고    scopus 로고
    • Glutathione conjugates of 4-hydroxy-2(E)-nonenal as biomarkers of hepatic oxidative stress-induced lipid peroxidation in rats
    • DOI 10.1016/j.freeradbiomed.2005.02.015, PII S0891584905000870
    • Völkel W., Alvarez-Sánchez R., Weick I., Mally A., Dekant W., Pähler A., Glutathione conjugates of 4-hydroxy-2(E)-nonenal as biomarkers of hepatic oxidative stress-induced lipid peroxidation in rats. Free Radical Biology and Medicine 2005 38 11 1526 1536 2-s2.0-18844408406 10.1016/j.freeradbiomed.2005.02.015 (Pubitemid 40693837)
    • (2005) Free Radical Biology and Medicine , vol.38 , Issue.11 , pp. 1526-1536
    • Volkel, W.1    Alvarez-Sanchez, R.2    Weick, I.3    Mally, A.4    Dekant, W.5    Pahler, A.6
  • 434
    • 52649104870 scopus 로고    scopus 로고
    • Effect of melatonin, curcumin, quercetin, and resveratrol on acute ferric nitrilotriacetate (Fe-NTA)-induced renal oxidative damage in rat
    • 2-s2.0-52649104870 10.1177/0960327108094508
    • Eybl V., Kotyzová D., Černá P., Koutenský J., Effect of melatonin, curcumin, quercetin, and resveratrol on acute ferric nitrilotriacetate (Fe-NTA)-induced renal oxidative damage in rat. Human and Experimental Toxicology 2008 27 4 347 353 2-s2.0-52649104870 10.1177/0960327108094508
    • (2008) Human and Experimental Toxicology , vol.27 , Issue.4 , pp. 347-353
    • Eybl, V.1    Kotyzová, D.2    Černá, P.3    Koutenský, J.4


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.