Korean Circ J.  2016 Jan;46(1):1-12. 10.4070/kcj.2016.46.1.1.

Post-Translational Modifications of Cardiac Mitochondrial Proteins in Cardiovascular Disease: Not Lost in Translation

Affiliations
  • 1Department of Health Sciences and Technology, Graduate School of Inje University, Busan, Korea. phyhanj@inje.ac.kr
  • 2National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea.

Abstract

Protein post-translational modifications (PTMs) are crucial in regulating cellular biology by playing key roles in processes such as the rapid on and off switching of signaling network and the regulation of enzymatic activities without affecting gene expressions. PTMs lead to conformational changes in the tertiary structure of protein and resultant regulation of protein function such as activation, inhibition, or signaling roles. PTMs such as phosphorylation, acetylation, and S-nitrosylation of specific sites in proteins have key roles in regulation of mitochondrial functions, thereby contributing to the progression to heart failure. Despite the extensive study of PTMs in mitochondrial proteins much remains unclear. Further research is yet to be undertaken to elucidate how changes in the proteins may lead to cardiovascular and metabolic disease progression in particular. We aimed to summarize the various types of PTMs that occur in mitochondrial proteins, which might be associated with heart failure. This study will increase the understanding of cardiovascular diseases through PTM.

Keyword

Cardiovascular diseases; Heart failure; Mitochondria; Post-translational modifications

MeSH Terms

Acetylation
Cardiovascular Diseases*
Gene Expression
Heart Failure
Metabolic Diseases
Mitochondria
Mitochondrial Proteins*
Phosphorylation
Protein Processing, Post-Translational*
Mitochondrial Proteins

Figure

  • Fig. 1 Several types of post-translational modifications. (A) Reversible protein phosphorylation. Protein kinase transfers a phosphate group (P) from ATP (ADPP) to the target protein. A protein phosphatase is responsible for removing the phosphate group via hydrolysis. (B) Protein kinases mediate phosphorylation at various amino acid sites of serine, threonine and tyrosine side chains. In the example shown, PDH is a phosphorylated protein found in the mitochondrial matrix. PDH is a large complex made up of several units (E1, E2, and E3), which is responsible for catalyzing oxidative decarboxylation of pyruvate, to form acetyl-CoA. PDH is phosphprylated at Ser264, 271, and 204. (C) Reversible protein acetylation. In the given example, Sirtuin 3 (SIRT3) deacetylases the acetylated form of NDUFA9. (D) Redox-mediated S-nitrosylation occurs through the covalent reaction of nitric oxide (NO)-related species with a cysteine thiol group on the target protein. ATP: adenosine triphosphate, ADP: adenosine diphosphate, PDH: pyruvate dehydrogenase, Ser: serine, NDUFA9: NADH dehydrogenase subcomplex A9, NO: nitric oxide, CI: complex I, SH: thiol subunit, S: sulfur.


Cited by  1 articles

NecroX-5 exerts anti-inflammatory and anti-fibrotic effects via modulation of the TNFα/Dcn/TGFβ1/Smad2 pathway in hypoxia/reoxygenation-treated rat hearts
Vu Thi Thu, Hyoung Kyu Kim, Le Thanh Long, To Thanh Thuy, Nguyen Quang Huy, Soon Ha Kim, Nari Kim, Kyung Soo Ko, Byoung Doo Rhee, Jin Han
Korean J Physiol Pharmacol. 2016;20(3):305-314.    doi: 10.4196/kjpp.2016.20.3.305.


Reference

1. Grotenbreg G, Ploegh H. Chemical biology: dressed-up proteins. Nature. 2007; 446:993–995.
2. Geiss-Friedlander R, Melchior F. Concepts in sumoylation: a decade on. Nat Rev Mol Cell Biol. 2007; 8:947–956.
3. Morrison RS, Kinoshita Y, Johnson MD, et al. Proteomic analysis in the neurosciences. Mol Cell Proteomics. 2002; 1:553–560.
4. Fukuda H, Sano N, Muto S, Horikoshi M. Simple histone acetylation plays a complex role in the regulation of gene expression. Brief Funct Genomic Proteomic. 2006; 5:190–208.
5. Chu S, Ferro TJ. Sp1: regulation of gene expression by phosphorylation. Gene. 2005; 348:1–11.
6. Li X, Foley EA, Kawashima SA, et al. Examining post-translational modification-mediated protein-protein interactions using a chemical proteomics approach. Protein Sci. 2013; 22:287–295.
7. Chavez JD, Weisbrod CR, Zheng C, Eng JK, Bruce JE. Protein interactions, post-translational modifications and topologies in human cells. Mol Cell Proteomics. 2013; 12:1451–1467.
8. Wang S, Ionescu R, Peekhaus N, Leung JY, Ha S, Vlasak J. Separation of post-translational modifications in monoclonal antibodies by exploiting subtle conformational changes under mildly acidic conditions. J Chromatogr A. 2010; 1217:6496–6502.
9. Slade DJ, Subramanian V, Fuhrmann J, Thompson PR. Chemical and biological methods to detect post-translational modifications of arginine. Biopolymers. 2014; 101:133–143.
10. Warnecke A, Sandalova T, Achour A, Harris RA. PyTMs: a useful PyMOL plugin for modeling common post-translational modifications. BMC Bioinformatics. 2014; 15:370.
11. Prabakaran S, Lippens G, Steen H, Gunawardena J. Post-translational modification: nature's escape from genetic imprisonment and the basis for dynamic information encoding. Wiley Interdiscip Rev Syst Biol Med. 2012; 4:565–583.
12. Liddy KA, White MY, Cordwell SJ. Functional decorations: posttranslational modifications and heart disease delineated by targeted proteomics. Genome Med. 2013; 5:20.
13. Han SJ, Lonard DM, O'Malley BW. Multi-modulation of nuclear receptor coactivators through posttranslational modifications. Trends Endocrinol Metab. 2009; 20:8–15.
14. Lonard DM, O'Malley BW. Nuclear receptor coregulators: judges, juries, and executioners of cellular regulation. Mol Cell. 2007; 27:691–700.
15. Rosenfeld MG, Lunyak VV, Glass CK. Sensors and signals: a coactivator/corepressor/epigenetic code for integrating signaldependent programs of transcriptional response. Genes Dev. 2006; 20:1405–1428.
16. Ren RJ, Dammer EB, Wang G, Seyfried NT, Levey AI. Proteomics of protein post-translational modifications implicated in neurodegeneration. Transl Neurodegener. 2014; 3:23.
17. Kemper JK. Regulation of FXR transcriptional activity in health and disease: Emerging roles of FXR cofactors and post-translational modifications. Biochim Biophys Acta. 2011; 1812:842–850.
18. Butkinaree C, Park K, Hart GW. O-linked beta-N-acetylglucosamine (O-GlcNAc): extensive crosstalk with phosphorylation to regulate signaling and transcription in response to nutrients and stress. Biochim Biophys Acta. 2010; 1800:96–106.
19. Ito K. Impact of post-translational modifications of proteins on the inflammatory process. Biochem Soc Trans. 2007; 35:281–283.
20. Moreno-Gonzalo O, Villarroya-Beltri C, Sánchez-Madrid F. Post-translational modifications of exosomal proteins. Front Immunol. 2014; 5:383.
21. Pejaver V, Hsu WL, Xin F, Dunker AK, Uversky VN, Radivojac P. The structural and functional signatures of proteins that undergo multiple events of post-translational modification. Protein Sci. 2014; 23:1077–1093.
22. Peng M, Scholten A, Heck AJ, van Breukelen B. Identification of enriched PTM crosstalk motifs from large-scale experimental data sets. J Proteome Res. 2014; 13:249–259.
23. Koc EC, Koc H. Regulation of mammalian mitochondrial translation by post-translational modifications. Biochim Biophys Acta. 2012; 1819:1055–1066.
24. Zhang J, Lin A, Powers J, et al. Perspectives on: SGP symposium on mitochondrial physiology and medicine: mitochondrial proteome design: from molecular identity to pathophysiological regulation. J Gen Physiol. 2012; 139:395–406.
25. Deng N, Zhang J, Zong C, et al. Phosphoproteome analysis reveals regulatory sites in major pathways of cardiac mitochondria. Mol Cell Proteomics. 2011; 10:M110.000117.
26. Papanicolaou KN, O'Rourke B, Foster DB. Metabolism leaves its mark on the powerhouse: recent progress in post-translational modifications of lysine in mitochondria. Front Physiol. 2014; 5:301.
27. Zhang Z, Tan M, Xie Z, Dai L, Chen Y, Zhao Y. Identification of lysine succinylation as a new post-translational modification. Nat Chem Biol. 2011; 7:58–63.
28. Hart GW, Slawson C, Ramirez-Correa G, Lagerlof O. Cross talk between O-GlcNAcylation and phosphorylation: roles in signaling, transcription, and chronic disease. Annu Rev Biochem. 2011; 80:825–858.
29. Gucek M, Murphy E. What can we learn about cardioprotection from the cardiac mitochondrial proteome? Cardiovasc Res. 2010; 88:211–218.
30. Narayan N, Lee IH, Borenstein R, et al. The NAD-dependent deacetylase SIRT2 is required for programmed necrosis. Nature. 2012; 492:199–204.
31. Hollander JM, Baseler WA, Dabkowski ER. Proteomic remodeling of mitochondria in heart failure. Congest Heart Fail. 2011; 17:262–268.
32. Nabel EG, Braunwald E. A tale of coronary artery disease and myocardial infarction. N Engl J Med. 2012; 366:54–63.
33. Chen J, Normand SL, Wang Y, Krumholz HM. National and regional trends in heart failure hospitalization and mortality rates for Medicare beneficiaries, 1998-2008. JAMA. 2011; 306:1669–1678.
34. Jhund PS, Macintyre K, Simpson CR, et al. Long-term trends in first hospitalization for heart failure and subsequent survival between 1986 and 2003: a population study of 5.1 million people. Circulation. 2009; 119:515–523.
35. Lee HA, Park H. Trends in ischemic heart disease mortality in Korea, 1985-2009: an age-period-cohort analysis. J Prev Med Public Health. 2012; 45:323–328.
36. Park JJ, Choi DJ. Treatment of heart failure with reduced ejection fraction: current update. Korean J Med. 2015; 88:127–134.
37. Rosca MG, Hoppel CL. Mitochondrial dysfunction in heart failure. Heart Fail Rev. 2013; 18:607–622.
38. White MY, Edwards AV, Cordwell SJ, Van Eyk JE. Mitochondria: a mirror into cellular dysfunction in heart disease. Proteomics Clin Appl. 2008; 2:845–861.
39. Haq S, Choukroun G, Lim H, et al. Differential activation of signal transduction pathways in human hearts with hypertrophy versus advanced heart failure. Circulation. 2001; 103:670–677.
40. De Sousa E, Veksler V, Minajeva A, et al. Subcellular creatine kinase alterations. Implications in heart failure. Circ Res. 1999; 85:68–76.
41. van der Velden J, Papp Z, Zaremba R, et al. Increased Ca2+-sensitivity of the contractile apparatus in end-stage human heart failure results from altered phosphorylation of contractile proteins. Cardiovasc Res. 2003; 57:37–47.
42. Dai DF, Rabinovitch PS, Ungvari Z. Mitochondria and cardiovascular aging. Circ Res. 2012; 110:1109–1124.
43. Boudina S, Laclau MN, Tariosse L, et al. Alteration of mitochondrial function in a model of chronic ischemia in vivo in rat heart. Am J Physiol Heart Circ Physiol. 2002; 282:H821–H831.
44. Sack MN, Rader TA, Park S, Bastin J, McCune SA, Kelly DP. Fatty acid oxidation enzyme gene expression is downregulated in the failing heart. Circulation. 1996; 94:2837–2842.
45. Kim SC, Sprung R, Chen Y, et al. Substrate and functional diversity of lysine acetylation revealed by a proteomics survey. Mol Cell. 2006; 23:607–618.
46. Choudhary C, Kumar C, Gnad F, et al. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science. 2009; 325:834–840.
47. Wagner GR, Payne RM. Mitochondrial acetylation and diseases of aging. J Aging Res. 2011; 2011:234875.
48. O'Rourke B, Van Eyk JE, Foster DB. Mitochondrial protein phosphorylation as a regulatory modality: implications for mitochondrial dysfunction in heart failure. Congest Heart Fail. 2011; 17:269–282.
49. Bayeva M, Gheorghiade M, Ardehali H. Mitochondria as a therapeutic target in heart failure. J Am Coll Cardiol. 2013; 61:599–610.
50. Kurdi M, Booz GW. Focus on mitochondria dysfunction and dysregulation in heart failure: towards new therapeutic strategies to improve heart function. Congest Heart Fail. 2011; 17:255–256.
51. Rosca M, Minkler P, Hoppel CL. Cardiac mitochondria in heart failure: normal cardiolipin profile and increased threonine phosphorylation of complex IV. Biochim Biophys Acta. 2011; 1807:1373–1382.
52. Karamanlidis G, Lee CF, Garcia-Menendez L, et al. Mitochondrial complex I deficiency increases protein acetylation and accelerates heart failure. Cell Metab. 2013; 18:239–250.
53. Hughes WA, Halestrap AP. The regulation of branched-chain 2-oxo acid dehydrogenase of liver, kidney and heart by phosphorylation. Biochem J. 1981; 196:459–469.
54. Johnson LN. The regulation of protein phosphorylation. Biochem Soc Trans. 2009; 37(Pt 4):627–641.
55. Hanks SK, Hunter T. Protein kinases 6. The eukaryotic protein kinase superfamily: kinase (catalytic) domain structure and classification. FASEB J. 1995; 9:576–596.
56. Barford D. Protein phosphatases. Curr Opin Struct Biol. 1995; 5:728–734.
57. Pagliarini DJ, Dixon JE. Mitochondrial modulation: reversible phosphorylation takes center stage? Trends Biochem Sci. 2006; 31:26–34.
58. Sun W, Liu Q, Leng J, Zheng Y, Li J. The role of Pyruvate Dehydrogenase Complex in cardiovascular diseases. Life Sci. 2015; 121:97–103.
59. Strumilo S. Short-term regulation of the mammalian pyruvate dehydrogenase complex. Acta Biochim Pol. 2005; 52:759–764.
60. Patel MS, Korotchkina LG. Regulation of the pyruvate dehydrogenase complex. Biochem Soc Trans. 2006; 34(Pt 2):217–222.
61. Gudi R, Bowker-Kinley MM, Kedishvili NY, Zhao Y, Popov KM. Diversity of the pyruvate dehydrogenase kinase gene family in humans. J Biol Chem. 1995; 270:28989–28994.
62. Patel MS, Korotchkina LG. Regulation of mammalian pyruvate dehydrogenase complex by phosphorylation: complexity of multiple phosphorylation sites and kinases. Exp Mol Med. 2001; 33:191–197.
63. Popov KM, Kedishvili NY, Zhao Y, Shimomura Y, Crabb DW, Harris RA. Primary structure of pyruvate dehydrogenase kinase establishes a new family of eukaryotic protein kinases. J Biol Chem. 1993; 268:26602–26606.
64. Kennelly PJ, Potts M. Fancy meeting you here! A fresh look at "prokaryotic" protein phosphorylation. J Bacteriol. 1996; 178:4759–4764.
65. Wolanin PM, Thomason PA, Stock JB. Histidine protein kinases: key signal transducers outside the animal kingdom. Genome Biol. 2002; 3:REVIEWS3013.1–REVIEWS3013.8.
66. Baker JC, Yan X, Peng T, Kasten S, Roche TE. Marked differences between two isoforms of human pyruvate dehydrogenase kinase. J Biol Chem. 2000; 275:15773–15781.
67. Bowker-Kinley MM, Davis WI, Wu P, Harris RA, Popov KM. Evidence for existence of tissue-specific regulation of the mammalian pyruvate dehydrogenase complex. Biochem J. 1998; 329(Pt 1):191–196.
68. Sugden MC, Holness MJ. Recent advances in mechanisms regulating glucose oxidation at the level of the pyruvate dehydrogenase complex by PDKs. Am J Physiol Endocrinol Metab. 2003; 284:E855–E862.
69. Roche TE, Hiromasa Y, Turkan A, et al. Essential roles of lipoyl domains in the activated function and control of pyruvate dehydrogenase kinases and phosphatase isoform 1. Eur J Biochem. 2003; 270:1050–1056.
70. Whitehouse S, Cooper RH, Randle PJ. Mechanism of activation of pyruvate dehydrogenase by dichloroacetate and other halogenated carboxylic acids. Biochem J. 1974; 141:761–774.
71. Xu M, Nagasaki M, Obayashi M, Sato Y, Tamura T, Shimomura Y. Mechanism of activation of branched-chain alpha-keto acid dehydrogenase complex by exercise. Biochem Biophys Res Commun. 2001; 287:752–756.
72. Zhang B, Zhao Y, Harris RA, Crabb DW. Molecular defects in the E1 alpha subunit of the branched-chain alpha-ketoacid dehydrogenase complex that cause maple syrup urine disease. Mol Biol Med. 1991; 8:39–47.
73. Schmidt U, Hajjar RJ, Kim CS, Lebeche D, Doye AA, Gwathmey JK. Human heart failure: cAMP stimulation of SR Ca(2+)-ATPase activity and phosphorylation level of phospholamban. Am J Physiol. 1999; 277(2 Pt 2):H474–H480.
74. Scacco S, Vergari R, Scarpulla RC, et al. cAMP-dependent phosphorylation of the nuclear encoded 18-kDa (IP) subunit of respiratory complex I and activation of the complex in serum-starved mouse fibroblast cultures. J Biol Chem. 2000; 275:17578–17582.
75. Papa S, De Rasmo D, Scacco S, et al. Mammalian complex I: a regulable and vulnerable pacemaker in mitochondrial respiratory function. Biochim Biophys Acta. 2008; 1777:719–728.
76. Palmisano G, Sardanelli AM, Signorile A, Papa S, Larsen MR. The phosphorylation pattern of bovine heart complex I subunits. Proteomics. 2007; 7:1575–1583.
77. Fang JK, Prabu SK, Sepuri NB, et al. Site specific phosphorylation of cytochrome c oxidase subunits I, IVi1 and Vb in rabbit hearts subjected to ischemia/reperfusion. FEBS Lett. 2007; 581:1302–1310.
78. Prabu SK, Anandatheerthavarada HK, Raza H, Srinivasan S, Spear JF, Avadhani NG. Protein kinase A-mediated phosphorylation modulates cytochrome c oxidase function and augments hypoxia and myocardial ischemia-related injury. J Biol Chem. 2006; 281:2061–2070.
79. Lee S. Post-translational modification of proteins in toxicological research: focus on lysine acylation. Toxicol Res. 2013; 29:81–86.
80. Sack MN, Finkel T. Mitochondrial metabolism, sirtuins, and aging. Cold Spring Harb Perspect Biol. 2012; (4):pii: a013102.
81. Anderson KA, Hirschey MD. Mitochondrial protein acetylation regulates metabolism. Essays Biochem. 2012; 52:23–35.
82. Hallows WC, Lee S, Denu JM. Sirtuins deacetylate and activate mammalian acetyl-CoA synthetases. Proc Natl Acad Sci U S A. 2006; 103:10230–10235.
83. Schwer B, Bunkenborg J, Verdin RO, Andersen JS, Verdin E. Reversible lysine acetylation controls the activity of the mitochondrial enzyme acetyl-CoA synthetase 2. Proc Natl Acad Sci U S A. 2006; 103:10224–10229.
84. Sundaresan NR, Samant SA, Pillai VB, Rajamohan SB, Gupta MP. SIRT3 is a stress-responsive deacetylase in cardiomyocytes that protects cells from stress-mediated cell death by deacetylation of Ku70. Mol Cell Biol. 2008; 28:6384–6401.
85. Sundaresan NR, Gupta M, Kim G, Rajamohan SB, Isbatan A, Gupta MP. Sirt3 blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent antioxidant defense mechanisms in mice. J Clin Invest. 2009; 119:2758–2771.
86. Hirschey MD, Shimazu T, Goetzman E, et al. SIRT3 regulates mitochondrial fatty-acid oxidation by reversible enzyme deacetylation. Nature. 2010; 464:121–125.
87. Zhao S, Xu W, Jiang W, et al. Regulation of cellular metabolism by protein lysine acetylation. Science. 2010; 327:1000–1004.
88. Schlicker C, Gertz M, Papatheodorou P, Kachholz B, Becker CF, Steegborn C. Substrates and regulation mechanisms for the human mitochondrial sirtuins Sirt3 and Sirt5. J Mol Biol. 2008; 382:790–801.
89. Ghanta S, Grossmann RE, Brenner C. Mitochondrial protein acetylation as a cell-intrinsic, evolutionary driver of fat storage: chemical and metabolic logic of acetyl-lysine modifications. Crit Rev Biochem Mol Biol. 2013; 48:561–574.
90. Lombard DB, Alt FW, Cheng HL, et al. Mammalian Sir2 homolog SIRT3 regulates global mitochondrial lysine acetylation. Mol Cell Biol. 2007; 27:8807–8814.
91. Mathias RA, Greco TM, Oberstein A, et al. Sirtuin 4 is a lipoamidase regulating pyruvate dehydrogenase complex activity. Cell. 2014; 159:1615–1625.
92. Hess DT, Matsumoto A, Kim SO, Marshall HE, Stamler JS. Protein S-nitrosylation: purview and parameters. Nat Rev Mol Cell Biol. 2005; 6:150–166.
93. Benhar M, Forrester MT, Hess DT, Stamler JS. Regulated protein denitrosylation by cytosolic and mitochondrial thioredoxins. Science. 2008; 320:1050–1054.
94. Sengupta R, Ryter SW, Zuckerbraun BS, Tzeng E, Billiar TR, Stoyanovsky DA. Thioredoxin catalyzes the denitrosation of low-molecular mass and protein S-nitrosothiols. Biochemistry. 2007; 46:8472–8483.
95. Moon KH, Kim BJ, Song BJ. Inhibition of mitochondrial aldehyde dehydrogenase by nitric oxide-mediated S-nitrosylation. FEBS Lett. 2005; 579:6115–6120.
96. Wolosker H, Panizzutti R, Engelender S. Inhibition of creatine kinase by S-nitrosoglutathione. FEBS Lett. 1996; 392:274–276.
97. Arstall MA, Bailey C, Gross WL, Bak M, Balligand JL, Kelly RA. Reversible S-nitrosation of creatine kinase by nitric oxide in adult rat ventricular myocytes. J Mol Cell Cardiol. 1998; 30:979–988.
98. Zhang J, Jin B, Li L, Block ER, Patel JM. Nitric oxide-induced persistent inhibition and nitrosylation of active site cysteine residues of mitochondrial cytochrome-c oxidase in lung endothelial cells. Am J Physiol Cell Physiol. 2005; 288:C840–C849.
99. Costa NJ, Dahm CC, Hurrell F, Taylor ER, Murphy MP. Interactions of mitochondrial thiols with nitric oxide. Antioxid Redox Signal. 2003; 5:291–305.
100. Zhang Y, Hogg N. S-Nitrosothiols: cellular formation and transport. Free Radic Biol Med. 2005; 38:831–838.
101. Lee JS, Smith E, Shilatifard A. The language of histone crosstalk. Cell. 2010; 142:682–685.
102. Seo J, Lee KJ. Post-translational modifications and their biological functions: proteomic analysis and systematic approaches. J Biochem Mol Biol. 2004; 37:35–44.
103. Ramirez-Correa GA, Ma J, Slawson C, et al. Removal of abnormal myofilament O-GlcNAcylation restores Ca2+ sensitivity in diabetic cardiac muscle. Diabetes. 2015; 64:3573–3587.
104. Matic I, Schimmel J, Hendriks IA, et al. Site-specific identification of SUMO-2 targets in cells reveals an inverted SUMOylation motif and a hydrophobic cluster SUMOylation motif. Mol Cell. 2010; 39:641–652.
105. Yao Q, Li H, Liu BQ, Huang XY, Guo L. SUMOylation-regulated protein phosphorylation, evidence from quantitative phosphoproteomics analyses. J Biol Chem. 2011; 286:27342–27349.
106. Rardin MJ, He W, Nishida Y, et al. SIRT5 regulates the mitochondrial lysine succinylome and metabolic networks. Cell Metab. 2013; 18:920–933.
107. Beer SM, Taylor ER, Brown SE, et al. Glutaredoxin 2 catalyzes the reversible oxidation and glutathionylation of mitochondrial membrane thiol proteins: implications for mitochondrial redox regulation and antioxidant DEFENSE. J Biol Chem. 2004; 279:47939–47951.
108. Hurd TR, Costa NJ, Dahm CC, et al. Glutathionylation of mitochondrial proteins. Antioxid Redox Signal. 2005; 7:999–1010.
109. Hurd TR, Filipovska A, Costa NJ, Dahm CC, Murphy MP. Disulphide formation on mitochondrial protein thiols. Biochem Soc Trans. 2005; 33(Pt 6):1390–1393.
110. Chen YR, Chen CL, Pfeiffer DR, Zweier JL. Mitochondrial complex II in the post-ischemic heart: oxidative injury and the role of protein S-glutathionylation. J Biol Chem. 2007; 282:32640–32654.
111. Hofer A, Wenz T. Post-translational modification of mitochondria as a novel mode of regulation. Exp Gerontol. 2014; 56:202–220.
112. Okazaki S, Yokoyama T, Miyauchi K, et al. Early statin treatment in patients with acute coronary syndrome: demonstration of the beneficial effect on atherosclerotic lesions by serial volumetric intravascular ultrasound analysis during half a year after coronary event: the ESTABLISH study. Circulation. 2004; 110:1061–1068.
113. Parang P, Singh B, Arora R. Metabolic modulators for chronic cardiac ischemia. J Cardiovasc Pharmacol Ther. 2005; 10:217–223.
114. Morrison DA, Sethi G, Sacks J, et al. Percutaneous coronary intervention versus coronary artery bypass graft surgery for patients with medically refractory myocardial ischemia and risk factors for adverse outcomes with bypass: a multicenter, randomized trial. Investigators of the Department of Veterans Affairs Cooperative Study #385, the Angina With Extremely Serious Operative Mortality Evaluation (AWESOME). J Am Coll Cardiol. 2001; 38:143–149.
115. Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation. 1986; 74:1124–1136.
116. Iliodromitis EK, Lazou A, Kremastinos DT. Ischemic preconditioning: protection against myocardial necrosis and apoptosis. Vasc Health Risk Manag. 2007; 3:629–637.
117. Kalogeris T, Bao Y, Korthuis RJ. Mitochondrial reactive oxygen species: a double edged sword in ischemia/reperfusion vs preconditioning. Redox Biol. 2014; 2:702–714.
118. Halestrap AP, Brenner C. The adenine nucleotide translocase: a central component of the mitochondrial permeability transition pore and key player in cell death. Curr Med Chem. 2003; 10:1507–1525.
119. Baines CP, Kaiser RA, Purcell NH, et al. Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature. 2005; 434:658–662.
120. Javadov S, Karmazyn M, Escobales N. Mitochondrial permeability transition pore opening as a promising therapeutic target in cardiac diseases. J Pharmacol Exp Ther. 2009; 330:670–678.
121. Smeele KM, Southworth R, Wu R, et al. Disruption of hexokinase IImitochondrial binding blocks ischemic preconditioning and causes rapid cardiac necrosis. Circ Res. 2011; 108:1165–1169.
122. McEnery MW, Snowman AM, Trifiletti RR, Snyder SH. Isolation of the mitochondrial benzodiazepine receptor: association with the voltage-dependent anion channel and the adenine nucleotide carrier. Proc Natl Acad Sci U S A. 1992; 89:3170–3174.
123. Onishi A, Miyamae M, Kaneda K, Kotani J, Figueredo VM. Direct evidence for inhibition of mitochondrial permeability transition pore opening by sevoflurane preconditioning in cardiomyocytes: comparison with cyclosporine A. Eur J Pharmacol. 2012; 675:40–46.
124. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N Engl J Med. 1993; 329:977–986.
125. Wende AR. Post-translational modifications of the cardiac proteome in diabetes and heart failure. Proteomics Clin Appl. 2016; [Epub ahead of print].
126. Park KJ, Kim YJ, Kim J, et al. Protective effects of peroxiredoxin on hydrogen peroxide induced oxidative stress and apoptosis in cardiomyocytes. Korean Circ J. 2012; 42:23–32.
127. Akhmedov D, De Marchi U, Wollheim CB, Wiederkehr A. Pyruvate dehydrogenase E1α phosphorylation is induced by glucose but does not control metabolism-secretion coupling in INS-1E clonal β-cells. Biochim Biophys Acta. 2012; 1823:1815–1824.
128. Kolobova E, Tuganova A, Boulatnikov I, Popov KM. Regulation of pyruvate dehydrogenase activity through phosphorylation at multiple sites. Biochem J. 2001; 358(Pt 1):69–77.
129. Churchill EN, Murriel CL, Chen CH, Mochly-Rosen D, Szweda LI. Reperfusion-induced translocation of deltaPKC to cardiac mitochondria prevents pyruvate dehydrogenase reactivation. Circ Res. 2005; 97:78–85.
130. Li H, Ren Z, Kang X, et al. Identification of tyrosine-phosphorylated proteins associated with metastasis and functional analysis of FER in human hepatocellular carcinoma cells. BMC Cancer. 2009; 9:366.
131. Choudhary C, Olsen JV, Brandts C, et al. Mislocalized activation of oncogenic RTKs switches downstream signaling outcomes. Mol Cell. 2009; 36:326–339.
132. Sharma K, D'Souza RC, Tyanova S, et al. Ultradeep human phosphoproteome reveals a distinct regulatory nature of Tyr and Ser/Thr-based signaling. Cell Rep. 2014; 8:1583–1594.
133. Zhao X, León IR, Bak S, et al. Phosphoproteome analysis of functional mitochondria isolated from resting human muscle reveals extensive phosphorylation of inner membrane protein complexes and enzymes. Mol Cell Proteomics. 2011; 10:M110.000299.
134. Grimsrud PA, Carson JJ, Hebert AS, et al. A quantitative map of the liver mitochondrial phosphoproteome reveals posttranslational control of ketogenesis. Cell Metab. 2012; 16:672–683.
135. Chen R, Fearnley IM, Peak-Chew SY, Walker JE. The phosphorylation of subunits of complex I from bovine heart mitochondria. J Biol Chem. 2004; 279:26036–26045.
136. Wu CC, MacCoss MJ, Howell KE, Yates JR 3rd. A method for the comprehensive proteomic analysis of membrane proteins. Nat Biotechnol. 2003; 21:532–538.
137. Raha S, Myint AT, Johnstone L, Robinson BH. Control of oxygen free radical formation from mitochondrial complex I: roles for protein kinase A and pyruvate dehydrogenase kinase. Free Radic Biol Med. 2002; 32:421–430.
138. Lee I, Salomon AR, Ficarro S, et al. cAMP-dependent tyrosine phosphorylation of subunit I inhibits cytochrome c oxidase activity. J Biol Chem. 2005; 280:6094–6100.
139. Steenaart NA, Shore GC. Mitochondrial cytochrome c oxidase subunit IV is phosphorylated by an endogenous kinase. FEBS Lett. 1997; 415:294–298.
140. Højlund K, Wrzesinski K, Larsen PM, et al. Proteome analysis reveals phosphorylation of ATP synthase beta -subunit in human skeletal muscle and proteins with potential roles in type 2 diabetes. J Biol Chem. 2003; 278:10436–10442.
141. Phillips D, Covian R, Aponte AM, et al. Regulation of oxidative phosphorylation complex activity: effects of tissue-specific metabolic stress within an allometric series and acute changes in workload. Am J Physiol Regul Integr Comp Physiol. 2012; 302:R1034–R1048.
142. Schulenberg B, Aggeler R, Beechem JM, Capaldi RA, Patton WF. Analysis of steady-state protein phosphorylation in mitochondria using a novel fluorescent phosphosensor dye. J Biol Chem. 2003; 278:27251–27255.
143. Bian Y, Song C, Cheng K, et al. An enzyme assisted RP-RPLC approach for in-depth analysis of human liver phosphoproteome. J Proteomics. 2014; 96:253–262.
144. Kettenbach AN, Schweppe DK, Faherty BK, Pechenick D, Pletnev AA, Gerber SA. Quantitative phosphoproteomics identifies substrates and functional modules of Aurora and Polo-like kinase activities in mitotic cells. Sci Signal. 2011; 4:rs5.
145. Huttlin EL, Jedrychowski MP, Elias JE, et al. A tissue-specific atlas of mouse protein phosphorylation and expression. Cell. 2010; 143:1174–1189.
146. Feng J, Lucchinetti E, Enkavi G, et al. Tyrosine phosphorylation by Src within the cavity of the adenine nucleotide translocase 1 regulates ADP/ATP exchange in mitochondria. Am J Physiol Cell Physiol. 2010; 298:C740–C748.
147. Wiśniewski JR, Nagaraj N, Zougman A, Gnad F, Mann M. Brain phosphoproteome obtained by a FASP-based method reveals plasma membrane protein topology. J Proteome Res. 2010; 9:3280–3289.
148. Xue L, Xu F, Meng L, et al. Acetylation-dependent regulation of mitochondrial ALDH2 activation by SIRT3 mediates acute ethanol-induced eNOS activation. FEBS Lett. 2012; 586:137–142.
149. Lu Z, Bourdi M, Li JH, et al. SIRT3-dependent deacetylation exacerbates acetaminophen hepatotoxicity. EMBO Rep. 2011; 12:840–846.
150. Shinmura K, Tamaki K, Sano M, et al. Caloric restriction primes mitochondria for ischemic stress by deacetylating specific mitochondrial proteins of the electron transport chain. Circ Res. 2011; 109:396–406.
151. Nguyen TT, Wong R, Menazza S, et al. Cyclophilin D modulates mitochondrial acetylome. Circ Res. 2013; 113:1308–1319.
152. Foster DB, Liu T, Rucker J, et al. The cardiac acetyl-lysine proteome. PLoS One. 2013; 8:e67513.
153. Grillon JM, Johnson KR, Kotlo K, Danziger RS. Non-histone lysine acetylated proteins in heart failure. Biochim Biophys Acta. 2012; 1822:607–614.
154. Li T, Liu M, Feng X, et al. Glyceraldehyde-3-phosphate dehydrogenase is activated by lysine 254 acetylation in response to glucose signal. J Biol Chem. 2014; 289:3775–3785.
155. Ahn BH, Kim HS, Song S, et al. A role for the mitochondrial deacetylase Sirt3 in regulating energy homeostasis. Proc Natl Acad Sci U S A. 2008; 105:14447–14452.
156. Qiu X, Brown K, Hirschey MD, Verdin E, Chen D. Calorie restriction reduces oxidative stress by SIRT3-mediated SOD2 activation. Cell Metab. 2010; 12:662–667.
Full Text Links
  • KCJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr