1932

Abstract

Oxidative injury due to elevated levels of reactive oxygen species is implicated in cardiovascular diseases, Alzheimer's disease, lung and liver diseases, and many cancers. Antioxidant therapies have generally been ineffective at treating these diseases, potentially due to ineffective doses but also due to interference with critical host defense and signaling processes. Therefore, alternative strategies to prevent oxidative injury are needed. Elevated levels of reactive oxygen species induce lipid peroxidation, generating reactive lipid dicarbonyls. These lipid oxidation products may be the most salient mediators of oxidative injury, as they cause cellular and organ dysfunction by adducting to proteins, lipids, and DNA. Small-molecule compounds have been developed in the past decade to selectively and effectively scavenge these reactive lipid dicarbonyls. This review outlines evidence supporting the role of lipid dicarbonyls in disease pathogenesis, as well as preclinical data supporting the efficacy of novel dicarbonyl scavengers in treating or preventing disease.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pharmtox-031620-035348
2021-01-06
2024-04-26
Loading full text...

Full text loading...

/deliver/fulltext/pharmtox/61/1/annurev-pharmtox-031620-035348.html?itemId=/content/journals/10.1146/annurev-pharmtox-031620-035348&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Conti V, Izzo V, Corbi G, Russomanno G, Manzo V et al. 2016. Antioxidant supplementation in the treatment of aging-associated diseases. Front. Pharmacol. 7:24
    [Google Scholar]
  2. 2. 
    Clarke R, Armitage J. 2002. Antioxidant vitamins and risk of cardiovascular disease. Review of large-scale randomised trials. Cardiovasc. Drugs Ther. 16:411–15
    [Google Scholar]
  3. 3. 
    Steinhubl SR. 2008. Why have antioxidants failed in clinical trials. ? Am. J. Cardiol. 101:14D–19D
    [Google Scholar]
  4. 4. 
    Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, Gluud C 2007. Mortality in randomized trials of antioxidant supplements for primary and secondary prevention: systematic review and meta-analysis. JAMA 297:842–57
    [Google Scholar]
  5. 5. 
    Roberts LJ2nd, Oates JA, Linton MF, Fazio S, Meador BP et al. 2007. The relationship between dose of vitamin E and suppression of oxidative stress in humans. Free Radic. Biol. Med. 43:1388–93
    [Google Scholar]
  6. 6. 
    Dodd S, Dean O, Copolov DL, Malhi GS, Berk M 2008. N-acetylcysteine for antioxidant therapy: pharmacology and clinical utility. Expert Opin. Biol. Ther. 8:1955–62
    [Google Scholar]
  7. 7. 
    Chu C, Deng J, Man Y, Qu Y 2017. Green tea extracts epigallocatechin-3-gallate for different treatments. Biomed. Res. Int. 2017:5615647
    [Google Scholar]
  8. 8. 
    Salehi B, Stojanović-Radić Z, Matejić J, Sharifi-Rad M, Anil Kumar NV et al. 2019. The therapeutic potential of curcumin: a review of clinical trials. Eur. J. Med. Chem. 163:527–45
    [Google Scholar]
  9. 9. 
    Davies SS, May-Zhang LS, Boutaud O, Amarnath V, Kirabo A, Harrison DG 2020. Isolevuglandins as mediators of disease and the development of dicarbonyl scavengers as pharmaceutical interventions. Pharmacol. Ther. 205:107418
    [Google Scholar]
  10. 10. 
    Iyer RS, Ghosh S, Salomon RG 1989. Levuglandin E2 crosslinks proteins. Prostaglandins 37:471–80
    [Google Scholar]
  11. 11. 
    Boutaud O, Brame CJ, Salomon RG, Roberts LJ2nd, Oates JA 1999. Characterization of the lysyl adducts formed from prostaglandin H2 via the levuglandin pathway. Biochemistry 38:9389–96
    [Google Scholar]
  12. 12. 
    Jirousek MR, Murthi KK, Salomon RG 1990. Electrophilic levuglandin E2-protein adducts bind glycine: a model for protein crosslinking. Prostaglandins 40:187–203
    [Google Scholar]
  13. 13. 
    Chio KS, Tappel AL. 1969. Synthesis and characterization of the fluorescent products derived from malonaldehyde and amino acids. Biochemistry 8:2821–26
    [Google Scholar]
  14. 14. 
    Esterbauer H, Schaur RJ, Zollner H 1991. Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radic. Biol. Med. 11:81–128
    [Google Scholar]
  15. 15. 
    Slatter DA, Avery NC, Bailey AJ 2004. Identification of a new cross-link and unique histidine adduct from bovine serum albumin incubated with malondialdehyde. J. Biol. Chem. 279:61–69
    [Google Scholar]
  16. 16. 
    Shao B, Pennathur S, Pagani I, Oda MN, Witztum JL et al. 2010. Modifying apolipoprotein A-I by malondialdehyde, but not by an array of other reactive carbonyls, blocks cholesterol efflux by the ABCA1 pathway. J. Biol. Chem. 285:18473–84
    [Google Scholar]
  17. 17. 
    Davies SS, Zhang LS. 2017. Reactive carbonyl species scavengers—novel therapeutic approaches for chronic diseases. Curr. Pharmacol. Rep. 3:51–67
    [Google Scholar]
  18. 18. 
    Zhang W-H, Liu J, Xu G, Yuan Q, Sayre LM 2003. Model studies on protein side chain modification by 4-oxo-2-nonenal. Chem. Res. Toxicol. 16:512–23
    [Google Scholar]
  19. 19. 
    Galligan JJ, Rose KL, Beavers WN, Hill S, Tallman KA et al. 2014. Stable histone adduction by 4-oxo-2-nonenal: a potential link between oxidative stress and epigenetics. J. Am. Chem. Soc. 136:11864–66
    [Google Scholar]
  20. 20. 
    Amarnath V, Amarnath K, Amarnath K, Davies S, Roberts LJ2nd 2004. Pyridoxamine: an extremely potent scavenger of 1,4-dicarbonyls. Chem. Res. Toxicol. 17:410–15
    [Google Scholar]
  21. 21. 
    Caldés C, Vilanova B, Adrover M, Muñoz F, Donoso J 2011. Phenol group in pyridoxamine acts as a stabilizing element for its carbinolamines and Schiff bases. Chem. Biodivers. 8:1318–32
    [Google Scholar]
  22. 22. 
    Sayre LM, Arora PK, Iyer RS, Salomon RG 1993. Pyrrole formation from 4-hydroxynonenal and primary amines. Chem. Res. Toxicol. 6:19–22
    [Google Scholar]
  23. 23. 
    Davies SS, Brantley EJ, Voziyan PA, Amarnath V, Zagol-Ikapitte I et al. 2006. Pyridoxamine analogues scavenge lipid-derived γ-ketoaldehydes and protect against H2O2-mediated cytotoxicity. Biochemistry 45:15756–67
    [Google Scholar]
  24. 24. 
    Brame CJ, Boutaud O, Davies SS, Yang T, Oates JA et al. 2004. Modification of proteins by isoketal-containing oxidized phospholipids. J. Biol. Chem. 279:13447–51
    [Google Scholar]
  25. 25. 
    Zagol-Ikapitte I, Amarnath V, Bala M, Roberts LJ2nd, Oates JA, Boutaud O 2010. Characterization of scavengers of γ-ketoaldehydes that do not inhibit prostaglandin biosynthesis. Chem. Res. Toxicol. 23:240–50
    [Google Scholar]
  26. 26. 
    Mayorov V, Uchakin P, Amarnath V, Panov AV, Bridges CC et al. 2019. Targeting of reactive isolevuglandins in mitochondrial dysfunction and inflammation. Redox Biol 26:101300
    [Google Scholar]
  27. 27. 
    Pitchford LM, Smith JD, Abumrad NN, Rathmacher JA, Fuller JC Jr 2018. Acute and 28-day repeated dose toxicity evaluations of 2-hydroxybenzylamine acetate in mice and rats. Regul. Toxicol. Pharmacol. 98:190–98
    [Google Scholar]
  28. 28. 
    Pitchford LM, Rathmacher JA, Fuller JC Jr, Daniels JS, Morrison RD et al. 2019. First-in-human study assessing safety, tolerability, and pharmacokinetics of 2-hydroxybenzylamine acetate, a selective dicarbonyl electrophile scavenger, in healthy volunteers. BMC Pharmacol. Toxicol. 20:1
    [Google Scholar]
  29. 29. 
    Pitchford LM, Driver PM, Fuller JC Jr, Akers WS, Abumrad NN et al. 2020. Safety, tolerability, and pharmacokinetics of repeated oral doses of 2-hydroxybenzylamine acetate in healthy volunteers: a double-blind, randomized, placebo-controlled clinical trial. BMC Pharmacol. Toxicol. 21:3
    [Google Scholar]
  30. 30. 
    Zagol-Ikapitte IA, Matafonova E, Amarnath V, Bodine CL, Boutaud O et al. 2010. Determination of the pharmacokinetics and oral bioavailability of salicylamine, a potent γ-ketoaldehyde scavenger, by LC/MS/MS. Pharmaceutics 2:18–29
    [Google Scholar]
  31. 31. 
    Kirabo A, Fontana V, de Faria AP, Loperena R, Galindo CL et al. 2014. DC isoketal-modified proteins activate T cells and promote hypertension. J. Clin. Investig. 124:4642–56
    [Google Scholar]
  32. 32. 
    Echt DS, Liebson PR, Mitchell LB, Peters RW, Obias-Manno D et al. 1991. Mortality and morbidity in patients receiving encainide, flecainide, or placebo. The Cardiac Arrhythmia Suppression Trial. N. Engl. J. Med. 324:781–88
    [Google Scholar]
  33. 33. 
    Akiyama T, Pawitan Y, Greenberg H, Kuo CS, Reynolds-Haertle RAet al 1991. Increased risk of death and cardiac arrest from encainide and flecainide in patients after non-Q-wave acute myocardial infarction in the Cardiac Arrhythmia Suppression Trial. Am. J. Cardiol. 68:1551–55
    [Google Scholar]
  34. 34. 
    Fukuda K, Davies SS, Nakajima T, Ong BH, Kupershmidt S et al. 2005. Oxidative mediated lipid peroxidation recapitulates proarrhythmic effects on cardiac sodium channels. Circ. Res. 97:1262–69
    [Google Scholar]
  35. 35. 
    Nakajima T, Davies SS, Matafonova E, Potet F, Amarnath V et al. 2010. Selective γ-ketoaldehyde scavengers protect NaV1.5 from oxidant-induced inactivation. J. Mol. Cell. Cardiol. 48:352–59
    [Google Scholar]
  36. 36. 
    Savelieva I, Kakouros N, Kourliouros A, Camm AJ 2011. Upstream therapies for management of atrial fibrillation: review of clinical evidence and implications for European Society of Cardiology guidelines. Part II: secondary prevention. Europace 13:610–25
    [Google Scholar]
  37. 37. 
    Boutaud O, Ou JJ, Chaurand P, Caprioli RM, Montine TJ, Oates JA 2002. Prostaglandin H2 (PGH2) accelerates formation of amyloid β1–42 oligomers. J. Neurochem. 82:1003–6
    [Google Scholar]
  38. 38. 
    He Y, Chen D, Huang PJ, Zhou Y, Ma L et al. 2018. Misfolding of a DNAzyme for ultrahigh sodium selectivity over potassium. Nucleic Acids Res 46:10262–71
    [Google Scholar]
  39. 39. 
    Guerrero-Muñoz MJ, Castillo-Carranza DL, Kayed R 2014. Therapeutic approaches against common structural features of toxic oligomers shared by multiple amyloidogenic proteins. Biochem. Pharmacol. 88:468–78
    [Google Scholar]
  40. 40. 
    Sidorova TN, Mace LC, Wells KS, Yermalitskaya LV, Su PF et al. 2014. Quantitative imaging of preamyloid oligomers, a novel structural abnormality, in human atrial samples. J. Histochem. Cytochem. 62:479–87
    [Google Scholar]
  41. 41. 
    Sidorova TN, Yermalitskaya LV, Mace LC, Wells KS, Boutaud O et al. 2015. Reactive γ-ketoaldehydes promote protein misfolding and preamyloid oligomer formation in rapidly-activated atrial cells. J. Mol. Cell. Cardiol. 79:295–302
    [Google Scholar]
  42. 42. 
    Prinsen JK, Kannankeril PJ, Sidorova TN, Yermalitskaya LV, Boutaud O et al. 2020. Highly reactive isolevuglandins promote atrial fibrillation caused by hypertension. JACC Basic Transl. Sci. 5:602–15
    [Google Scholar]
  43. 43. 
    Zagol-Ikapitte I, Masterson TS, Amarnath V, Montine TJ, Andreasson KI et al. 2005. Prostaglandin H2-derived adducts of proteins correlate with Alzheimer's disease severity. J. Neurochem. 94:1140–45
    [Google Scholar]
  44. 44. 
    Prinsen JK, Kannankeril PJ, Yermalitskaya LV, Jafarian-Kerman SR, Zagol-Ikapitte I et al. 2016. Highly-reactive isolevuglandins promote atrial fibrillation susceptibility in obesity. Circulation 134:Suppl. 1A20445
    [Google Scholar]
  45. 45. 
    Whelton PK, Carey RM, Aronow WS, Casey DE Jr, Collins KJ et al. 2017. ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 138:e426–83
    [Google Scholar]
  46. 46. 
    Guzik TJ, Hoch NE, Brown KA, McCann LA, Rahman A et al. 2007. Role of the T cell in the genesis of angiotensin II induced hypertension and vascular dysfunction. J. Exp. Med. 204:2449–60
    [Google Scholar]
  47. 47. 
    Crowley SD, Song Y-S, Lin EE, Griffiths R, Kim H-S, Ruiz P 2010. Lymphocyte responses exacerbate angiotensin II-dependent hypertension. Am. J. Physiol. Regul. Integr. Comp. Physiol. 298:R1089–97
    [Google Scholar]
  48. 48. 
    Madhur MS, Lob HE, McCann LA, Iwakura Y, Blinder Y et al. 2010. Interleukin 17 promotes angiotensin II-induced hypertension and vascular dysfunction. Hypertension 55:500–7
    [Google Scholar]
  49. 49. 
    Angel K, Provan SA, Fagerhol MK, Mowinckel P, Kvien TK, Atar D 2012. Effect of 1-year anti-TNF-α therapy on aortic stiffness, carotid atherosclerosis, and calprotectin in inflammatory arthropathies: a controlled study. Am. J. Hypertens. 25:644–50
    [Google Scholar]
  50. 50. 
    Angel K, Provan SA, Gulseth HL, Mowinckel P, Kvien TK, Atar D 2010. Tumor necrosis factor-α antagonists improve aortic stiffness in patients with inflammatory arthropathies: a controlled study. Hypertension 55:333–38
    [Google Scholar]
  51. 51. 
    Moreau KL, Deane KD, Meditz AL, Kohrt WM 2013. Tumor necrosis factor-α inhibition improves endothelial function and decreases arterial stiffness in estrogen-deficient postmenopausal women. Atherosclerosis 230:390–96
    [Google Scholar]
  52. 52. 
    Herrera J, Ferrebuz A, MacGregor EG, Rodriguez-Iturbe B 2006. Mycophenolate mofetil treatment improves hypertension in patients with psoriasis and rheumatoid arthritis. J. Am. Soc. Nephrol. 17:S218–25
    [Google Scholar]
  53. 53. 
    Vinh A, Chen W, Blinder Y, Weiss D, Taylor WR et al. 2010. Inhibition and genetic ablation of the B7/CD28 T-cell costimulation axis prevents experimental hypertension. Circulation 122:2529–37
    [Google Scholar]
  54. 54. 
    Itani HA, Xiao L, Saleh MA, Wu J, Pilkinton MA et al. 2016. CD70 exacerbates blood pressure elevation and renal damage in response to repeated hypertensive stimuli. Circ. Res. 118:1233–43
    [Google Scholar]
  55. 55. 
    Wu J, Saleh MA, Kirabo A, Itani HA, Montaniel KR et al. 2016. Immune activation caused by vascular oxidation promotes fibrosis and hypertension. J. Clin. Investig. 126:50–67
    [Google Scholar]
  56. 56. 
    Stavrovskaya IG, Baranov SV, Guo X, Davies SS, Roberts LJ 2nd, Kristal BS 2010. Reactive γ-ketoaldehydes formed via the isoprostane pathway disrupt mitochondrial respiration and calcium homeostasis. Free Radic. Biol. Med. 49:567–79
    [Google Scholar]
  57. 57. 
    Itani HA, Dikalova AE, McMaster WG, Nazarewicz RR, Bikineyeva AT et al. 2016. Mitochondrial cyclophilin D in vascular oxidative stress and hypertension. Hypertension 67:1218–27
    [Google Scholar]
  58. 58. 
    Linton MRF, Yancey PG, Davies SS, Jerome WG, Linton EF et al. 2019. The role of lipids and lipoproteins in atherosclerosis. Endotext Jan. 3. https://www.endotext.org/chapter/?p=6134
    [Google Scholar]
  59. 59. 
    Sampson UK, Fazio S, Linton MF 2012. Residual cardiovascular risk despite optimal LDL cholesterol reduction with statins: the evidence, etiology, and therapeutic challenges. Curr. Atheroscler. Rep. 14:1–10
    [Google Scholar]
  60. 60. 
    Nicholls SJ, Lincoff AM, Barter PJ, Brewer HB, Fox KA et al. 2015. Assessment of the clinical effects of cholesteryl ester transfer protein inhibition with evacetrapib in patients at high-risk for vascular outcomes: rationale and design of the ACCELERATE trial. Am. Heart J. 170:1061–69
    [Google Scholar]
  61. 61. 
    Hewing B, Fisher EA. 2012. Rationale for cholesteryl ester transfer protein inhibition. Curr. Opin. Lipidol. 23:372–76
    [Google Scholar]
  62. 62. 
    Kingwell BA, Chapman MJ, Kontush A, Miller NE 2014. HDL-targeted therapies: progress, failures and future. Nat. Rev. Drug Discov. 13:445–64
    [Google Scholar]
  63. 63. 
    Huang J, Wang D, Huang L-H, Huang H 2020. Roles of reconstituted high-density lipoprotein nanoparticles in cardiovascular disease: a new paradigm for drug discovery. Int. J. Mol. Sci. 21:3739
    [Google Scholar]
  64. 64. 
    Brownell N, Rohatgi A. 2016. Modulating cholesterol efflux capacity to improve cardiovascular disease. Curr. Opin. Lipidol. 27:398–407
    [Google Scholar]
  65. 65. 
    Bowry VW, Stanley KK, Stocker R 1992. High density lipoprotein is the major carrier of lipid hydroperoxides in human blood plasma from fasting donors. PNAS 89:10316–20
    [Google Scholar]
  66. 66. 
    Proudfoot JM, Barden AE, Loke WM, Croft KD, Puddey IB, Mori TA 2009. HDL is the major lipoprotein carrier of plasma F2-isoprostanes. J. Lipid Res. 50:716–22
    [Google Scholar]
  67. 67. 
    Milne GL, Yin H, Hardy KD, Davies SS, Roberts LJ 2nd 2011. Isoprostane generation and function. Chem. Rev. 111:5973–96
    [Google Scholar]
  68. 68. 
    Lawson JA, Rokach J, FitzGerald GA 1999. Isoprostanes: formation, analysis and use as indices of lipid peroxidation in vivo. J. Biol. Chem. 274:24441–44
    [Google Scholar]
  69. 69. 
    May-Zhang LS, Yermalitsky V, Huang J, Pleasent T, Borja MS et al. 2018. Modification by isolevuglandins, highly reactive γ-ketoaldehydes, deleteriously alters high-density lipoprotein structure and function. J. Biol. Chem. 293:9176–87
    [Google Scholar]
  70. 70. 
    May-Zhang LS, Yermalitsky V, Melchior JT, Morris J, Tallman KA et al. 2019. Modified sites and functional consequences of 4-oxo-2-nonenal adducts in HDL that are elevated in familial hypercholesterolemia. J. Biol. Chem. 294:19022–33
    [Google Scholar]
  71. 71. 
    Guo L, Chen Z, Amarnath V, Yancey PG, Van Lenten BJ et al. 2015. Isolevuglandin-type lipid aldehydes induce the inflammatory response of macrophages by modifying phosphatidylethanolamines and activating the receptor for advanced glycation endproducts. Antioxid. Redox Signal. 22:1633–45
    [Google Scholar]
  72. 72. 
    Tao H, Huang H, Yancey PG, Yermalitsky V, Blakemore JL et al. 2020. Scavenging of reactive dicarbonyls with 2-hydroxybenzylamine reduces atherosclerosis in hypercholesterolemic Ldlr−/− mice. Nat. Commun. 11:4084
    [Google Scholar]
  73. 73. 
    Huang J, Yancey PG, May-Zhang LS, Tao H, Zhang Y et al. 2019. Scavenging dicarbonyls with 5′-O-pentyl-pyridoxamine improves insulin sensitivity and reduces atherosclerosis through modulating inflammatory Ly6Chi monocytosis and macrophage polarization. bioRxiv 529339. https://doi.org/10.1101/529339
    [Crossref]
  74. 74. 
    Halliwell B. 2006. Oxidative stress and neurodegeneration: Where are we now. ? J. Neurochem. 97:1634–58
    [Google Scholar]
  75. 75. 
    Sultana R, Perluigi M, Butterfield DA 2013. Lipid peroxidation triggers neurodegeneration: a redox proteomics view into the Alzheimer disease brain. Free Radic. Biol. Med. 62:157–69
    [Google Scholar]
  76. 76. 
    Bonet-Costa V, Pomatto LC, Davies KJ 2016. The proteasome and oxidative stress in Alzheimer's disease. Antioxid. Redox Signal. 25:886–901
    [Google Scholar]
  77. 77. 
    Davies SS, Amarnath V, Montine KS, Bernoud-Hubac N, Boutaud O et al. 2002. Effects of reactive γ-ketoaldehydes formed by the isoprostane pathway (isoketals) and cyclooxygenase pathway (levuglandins) on proteasome function. FASEB J 16:715–17
    [Google Scholar]
  78. 78. 
    Boutaud O, Montine TJ, Chang L, Klein WL, Oates JA 2006. PGH2-derived levuglandin adducts increase the neurotoxicity of amyloid β1–42. J. Neurochem. 96:917–23
    [Google Scholar]
  79. 79. 
    Davies SS, Bodine C, Matafonova E, Pantazides BG, Bernoud-Hubac N et al. 2011. Treatment with a γ-ketoaldehyde scavenger prevents working memory deficits in hApoE4 mice. J. Alzheimers Dis. 27:49–59
    [Google Scholar]
  80. 80. 
    Shi J, Johansson J, Woodling NS, Wang Q, Montine TJ, Andreasson K 2010. The prostaglandin E2 E-prostanoid 4 receptor exerts anti-inflammatory effects in brain innate immunity. J. Immunol. 184:7207–18
    [Google Scholar]
  81. 81. 
    Liang X, Wang Q, Hand T, Wu L, Breyer RM et al. 2005. Deletion of the prostaglandin E2 EP2 receptor reduces oxidative damage and amyloid burden in a model of Alzheimer's disease. J. Neurosci. 25:10180–87
    [Google Scholar]
  82. 82. 
    Shi J, Wang Q, Johansson JU, Liang X, Woodling NS et al. 2012. Inflammatory prostaglandin E2 signaling in a mouse model of Alzheimer disease. Ann. Neurol. 72:788–98
    [Google Scholar]
  83. 83. 
    Zhen G, Kim YT, Li R-C, Yocum J, Kapoor N et al. 2012. PGE2 EP1 receptor exacerbated neurotoxicity in a mouse model of cerebral ischemia and Alzheimer's disease. Neurobiol. Aging 33:2215–19
    [Google Scholar]
  84. 84. 
    Woodling NS, Wang Q, Priyam PG, Larkin P, Shi J et al. 2014. Suppression of Alzheimer-associated inflammation by microglial prostaglandin-E2 EP4 receptor signaling. J. Neurosci. 34:5882–94
    [Google Scholar]
  85. 85. 
    Mont S, Davies SS, Roberts LJ 2nd, Mernaugh RL, McDonald WH et al. 2016. Accumulation of isolevuglandin-modified protein in normal and fibrotic lung. Sci. Rep. 6:24919
    [Google Scholar]
  86. 86. 
    Lane KL, Talati M, Austin E, Hemnes AR, Johnson JA et al. 2011. Oxidative injury is a common consequence of BMPR2 mutations. Pulm. Circ. 1:72–83
    [Google Scholar]
  87. 87. 
    Egnatchik RA, Brittain EL, Shah AT, Fares WH, Ford HJ et al. 2017. Dysfunctional BMPR2 signaling drives an abnormal endothelial requirement for glutamine in pulmonary arterial hypertension. Pulm. Circ. 7:186–99
    [Google Scholar]
  88. 88. 
    Xu F, Li M, Zhang C, Cui J, Liu J et al. 2017. Clinicopathological and prognostic significance of COX-2 immunohistochemical expression in breast cancer: a meta-analysis. Oncotarget 8:6003–12
    [Google Scholar]
  89. 89. 
    Shimizu K, Yukawa T, Okita R, Saisho S, Maeda A et al. 2015. Cyclooxygenase-2 expression is a prognostic biomarker for non-small cell lung cancer patients treated with adjuvant platinum-based chemotherapy. World J. Surg. Oncol. 13:21
    [Google Scholar]
  90. 90. 
    Masunaga R, Kohno H, Dhar DK, Ohno S, Shibakita M et al. 2000. Cyclooxygenase-2 expression correlates with tumor neovascularization and prognosis in human colorectal carcinoma patients. Clin. Cancer Res. 6:4064–68
    [Google Scholar]
  91. 91. 
    Carrier EJ, Zagol-Ikapitte I, Amarnath V, Boutaud O, Oates JA 2014. Levuglandin forms adducts with histone H4 in a cyclooxygenase-2-dependent manner, altering its interaction with DNA. Biochemistry 53:2436–41
    [Google Scholar]
  92. 92. 
    Farhadi A, Fields J, Banan A, Keshavarzian A 2002. Reactive oxygen species: Are they involved in the pathogenesis of GERD, Barrett's esophagus, and the latter's progression toward esophageal cancer. ? Am. J. Gastroenterol. 97:22–26
    [Google Scholar]
  93. 93. 
    Caspa Gokulan R, Adcock JM, Zagol-Ikapitte I, Mernaugh R, Williams P et al. 2019. Gastroesophageal reflux induces protein adducts in the esophagus. Cell. Mol. Gastroenterol. Hepatol. 7:480–82.e7
    [Google Scholar]
  94. 94. 
    Rouach H, Fataccioli V, Gentil M, French SW, Morimoto M, Nordmann R 1997. Effect of chronic ethanol feeding on lipid peroxidation and protein oxidation in relation to liver pathology. Hepatology 25:351–55
    [Google Scholar]
  95. 95. 
    Roychowdhury S, McMullen MR, Pritchard MT, Li W, Salomon RG, Nagy LE 2009. Formation of γ-ketoaldehyde-protein adducts during ethanol-induced liver injury in mice. Free Radic. Biol. Med. 47:1526–38
    [Google Scholar]
  96. 96. 
    Li W, Laird JM, Lu L, Roychowdhury S, Nagy LE et al. 2009. Isolevuglandins covalently modify phosphatidylethanolamines in vivo: detection and quantitative analysis of hydroxylactam adducts. Free Radic. Biol. Med. 47:1539–52
    [Google Scholar]
  97. 97. 
    Longato L, Andreola F, Davies SS, Roberts JL, Fusai G et al. 2017. Reactive γ-ketoaldehydes as novel activators of hepatic stellate cells in vitro. Free Radic. Biol. Med. 102:162–73
    [Google Scholar]
  98. 98. 
    Fuller JC Jr., Pitchford LM, Abumrad NN, Rathmacher JA. 2018. Subchronic (90-day) repeated dose oral toxicity study of 2-hydroxybenzylamine acetate in rabbit. Regul. Toxicol. Pharmacol. 100:52–58
    [Google Scholar]
  99. 99. 
    Fuller JC Jr., Pitchford LM, Abumrad NN, Rathmacher JA. 2018. Subchronic (90-day) repeated dose toxicity study of 2-hydroxybenzylamine acetate in rats. Regul. Toxicol. Pharmacol. 99:225–32
    [Google Scholar]
  100. 100. 
    Fuller JC Jr., Pitchford LM, Morrison RD, Daniels JS, Flynn CR et al. 2018. In vitro safety pharmacology evaluation of 2-hydroxybenzylamine acetate. Food Chem. Toxicol. 121:541–48
    [Google Scholar]
/content/journals/10.1146/annurev-pharmtox-031620-035348
Loading
/content/journals/10.1146/annurev-pharmtox-031620-035348
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error