1932

Abstract

Mitochondrial dysfunction is a key pathophysiological component of many acute and chronic diseases. Maintenance of mitochondrial homeostasis through the balance of mitochondrial turnover, fission and fusion, and generation of new mitochondria via mitochondrial biogenesis is critical for tissue health. Pharmacological activation of mitochondrial biogenesis can enhance oxidative metabolism and tissue bioenergetics, and improve organ function in conditions characterized by mitochondrial dysfunction. However, owing to the complexity of mitochondrial assembly and maintenance, identification of specific activators of mitochondrial biogenesis has been difficult. This review provides an overview of the role of mitochondrial dysfunction in acute and chronic diseases, details the current state of therapeutics for the stimulation of mitochondrial biogenesis and their effects on disease outcomes, describes new screening methodologies to identify novel stimulators and noncanonical pathways of mitochondrial biogenesis, and discusses potential hurdles of mitochondrial biogenesis as a therapeutic strategy.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pharmtox-010715-103155
2016-01-06
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/pharmtox/56/1/annurev-pharmtox-010715-103155.html?itemId=/content/journals/10.1146/annurev-pharmtox-010715-103155&mimeType=html&fmt=ahah

Literature Cited

  1. Chandel NS. 1.  2014. Mitochondria as signaling organelles. BMC Biol. 12:34 [Google Scholar]
  2. Kotiadis VN, Duchen MR, Osellame LD. 2.  2014. Mitochondrial quality control and communications with the nucleus are important in maintaining mitochondrial function and cell health. Biochim. Biophys. Acta 1840:1254–65 [Google Scholar]
  3. De Bock K, Georgiadou M, Carmeliet P. 3.  2013. Role of endothelial cell metabolism in vessel sprouting. Cell Metab. 18:634–47 [Google Scholar]
  4. Baker MJ, Tatsuta T, Langer T. 4.  2011. Quality control of mitochondrial proteostasis. Cold Spring Harb. Perspect. Biol. 3:a007559 [Google Scholar]
  5. Youle RJ, Narendra DP. 5.  2011. Mechanisms of mitophagy. Nat. Rev. Mol. Cell Biol. 12:9–14 [Google Scholar]
  6. Nunnari J, Suomalainen A. 6.  2012. Mitochondria: in sickness and in health. Cell 148:1145–59 [Google Scholar]
  7. Ishikawa K, Takenaga K, Akimoto M, Koshikawa N, Yamaguchi A. 7.  et al. 2008. ROS-generating mitochondrial DNA mutations can regulate tumor cell metastasis. Science 320:661–64 [Google Scholar]
  8. Cohen RA, Tong X. 8.  2010. Vascular oxidative stress: the common link in hypertensive and diabetic vascular disease. J. Cardiovasc. Pharmacol. 55:308–16 [Google Scholar]
  9. Andreux PA, Houtkooper RH, Auwerx J. 9.  2013. Pharmacological approaches to restore mitochondrial function. Nat. Rev. Drug Discov. 12:465–83 [Google Scholar]
  10. Stallons LJ, Funk JA, Schnellmann RG. 10.  2013. Mitochondrial homeostasis in acute organ failure. Curr. Pathobiol. Rep. 1:3169–77 [Google Scholar]
  11. Hausenloy DJ, Yellon DM. 11.  2013. Myocardial ischemia-reperfusion injury: a neglected therapeutic target. J. Clin. Investig. 123:92–100 [Google Scholar]
  12. Sanderson TH, Reynolds CA, Kumar R, Przyklenk K, Huttemann M. 12.  2013. Molecular mechanisms of ischemia-reperfusion injury in brain: pivotal role of the mitochondrial membrane potential in reactive oxygen species generation. Mol. Neurobiol. 47:9–23 [Google Scholar]
  13. McEwen ML, Sullivan PG, Rabchevsky AG, Springer JE. 13.  2011. Targeting mitochondrial function for the treatment of acute spinal cord injury. Neurotherapeutics 8:168–79 [Google Scholar]
  14. McGill MR, Sharpe MR, Williams CD, Taha M, Curry SC, Jaeschke H. 14.  2012. The mechanism underlying acetaminophen-induced hepatotoxicity in humans and mice involves mitochondrial damage and nuclear DNA fragmentation. J. Clin. Investig. 122:1574–83 [Google Scholar]
  15. Naguib YM, Azmy RM, Samaka RM, Salem MF. 15.  2014. Pleurotus ostreatus opposes mitochondrial dysfunction and oxidative stress in acetaminophen-induced hepato-renal injury. BMC Complement. Altern. Med. 14:494 [Google Scholar]
  16. Blake R, Trounce IA. 16.  2014. Mitochondrial dysfunction and complications associated with diabetes. Biochim. Biophys. Acta 1840:1404–12 [Google Scholar]
  17. Moreira PI, Carvalho C, Zhu X, Smith MA, Perry G. 17.  2010. Mitochondrial dysfunction is a trigger of Alzheimer's disease pathophysiology. Biochim. Biophys. Acta 1802:2–10 [Google Scholar]
  18. Chaturvedi RK, Beal MF. 18.  2013. Mitochondrial diseases of the brain. Free Radic. Biol. Med. 63:1–29 [Google Scholar]
  19. Chaturvedi RK, Beal MF. 19.  2013. Mitochondria targeted therapeutic approaches in Parkinson's and Huntington's diseases. Mol. Cell Neurosci. 55:101–14 [Google Scholar]
  20. Cozzolino M, Carri MT. 20.  2012. Mitochondrial dysfunction in ALS. Prog. Neurobiol. 97:54–66 [Google Scholar]
  21. Archer SL, Gomberg-Maitland M, Maitland ML, Rich S, Garcia JG, Weir EK. 21.  2008. Mitochondrial metabolism, redox signaling, and fusion: a mitochondria-ROS-HIF-1alpha-Kv1.5 O2-sensing pathway at the intersection of pulmonary hypertension and cancer. Am. J. Physiol. Heart Circ. Physiol. 294:H570–78 [Google Scholar]
  22. Bayeva M, Gheorghiade M, Ardehali H. 22.  2013. Mitochondria as a therapeutic target in heart failure. J. Am. Coll. Cardiol. 61:599–610 [Google Scholar]
  23. Sebastiani M, Giordano C, Nediani C, Travaglini C, Borchi E. 23.  et al. 2007. Induction of mitochondrial biogenesis is a maladaptive mechanism in mitochondrial cardiomyopathies. J. Am. Coll. Cardiol. 50:1362–69 [Google Scholar]
  24. Moskowitz MA, Lo EH, Iadecola C. 24.  2010. The science of stroke: mechanisms in search of treatments. Neuron 67:181–98 [Google Scholar]
  25. Kalogeris T, Bao Y, Korthuis RJ. 25.  2014. Mitochondrial reactive oxygen species: a double edged sword in ischemia/reperfusion versus preconditioning. Redox Biol. 2:702–14 [Google Scholar]
  26. Andreadou I, Iliodromitis EK, Farmakis D, Kremastinos DT. 26.  2009. To prevent, protect and save the ischemic heart: antioxidants revisited. Expert Opin. Ther. Targets 13:945–56 [Google Scholar]
  27. Adlam VJ, Harrison JC, Porteous CM, James AM, Smith RA. 27.  et al. 2005. Targeting an antioxidant to mitochondria decreases cardiac ischemia-reperfusion injury. FASEB J. 19:1088–95 [Google Scholar]
  28. Sun SY. 28.  2010. N-acetylcysteine, reactive oxygen species and beyond. Cancer Biol. Ther. 9:109–10 [Google Scholar]
  29. Skyschally A, Schulz R, Heusch G. 29.  2010. Cyclosporine A at reperfusion reduces infarct size in pigs. Cardiovasc. Drugs Ther. 24:85–87 [Google Scholar]
  30. Osman MM, Lulic D, Glover L, Stahl CE, Lau T. 30.  et al. 2011. Cyclosporine-A as a neuroprotective agent against stroke: its translation from laboratory research to clinical application. Neuropeptides 45:359–68 [Google Scholar]
  31. O'Connell S, Tuite N, Slattery C, Ryan MP, McMorrow T. 31.  2012. Cyclosporine A–induced oxidative stress in human renal mesangial cells: a role for ERK 1/2 MAPK signaling. Toxicol. Sci. 126:101–13 [Google Scholar]
  32. Jiang HK, Miao Y, Wang YH, Zhao M, Feng ZH. 32.  et al. 2014. Aerobic interval training protects against myocardial infarction-induced oxidative injury by enhancing antioxidase system and mitochondrial biosynthesis. Clin. Exp. Pharmacol. Physiol. 41:192–201 [Google Scholar]
  33. Ikeuchi M, Matsusaka H, Kang D, Matsushima S, Ide T. 33.  et al. 2005. Overexpression of mitochondrial transcription factor A ameliorates mitochondrial deficiencies and cardiac failure after myocardial infarction. Circulation 112:683–90 [Google Scholar]
  34. Hokari M, Kuroda S, Kinugawa S, Ide T, Tsutsui H, Iwasaki Y. 34.  2010. Overexpression of mitochondrial transcription factor A (TFAM) ameliorates delayed neuronal death due to transient forebrain ischemia in mice. Neuropathology 30:401–7 [Google Scholar]
  35. Jesinkey SR, Funk JA, Stallons LJ, Wills LP, Megyesi JK. 35.  et al. 2014. Formoterol restores mitochondrial and renal function after ischemia-reperfusion injury. J. Am. Soc. Nephrol. 25:1157–62 [Google Scholar]
  36. Whitaker RM, Wills LP, Stallons LJ, Schnellmann RG. 36.  2013. cGMP-selective phosphodiesterase inhibitors stimulate mitochondrial biogenesis and promote recovery from acute kidney injury. J. Pharmacol. Exp. Ther. 347:626–34 [Google Scholar]
  37. Garrett SM, Whitaker RM, Beeson CC, Schnellmann RG. 37.  2014. Agonism of the 5-hydroxytryptamine 1F receptor promotes mitochondrial biogenesis and recovery from acute kidney injury. J. Pharmacol. Exp. Ther. 350:257–64 [Google Scholar]
  38. Dam AD, Mitchell AS, Quadrilatero J. 38.  2013. Induction of mitochondrial biogenesis protects against caspase-dependent and caspase-independent apoptosis in L6 myoblasts. Biochim. Biophys. Acta 1833:3426–35 [Google Scholar]
  39. Che R, Yuan Y, Huang S, Zhang A. 39.  2014. Mitochondrial dysfunction in the pathophysiology of renal diseases. Am. J. Physiol. Ren. Physiol. 306:F367–78 [Google Scholar]
  40. Baker BM, Haynes CM. 40.  2011. Mitochondrial protein quality control during biogenesis and aging. Trends Biochem. Sci. 36:254–61 [Google Scholar]
  41. Baker MJ, Palmer CS, Stojanovski D. 41.  2014. Mitochondrial protein quality control in health and disease. Br. J. Pharmacol. 171:1870–89 [Google Scholar]
  42. Schapira AH. 42.  2008. Mitochondria in the aetiology and pathogenesis of Parkinson's disease. Lancet Neurol. 7:97–109 [Google Scholar]
  43. Bezard E, Yue Z, Kirik D, Spillantini MG. 43.  2013. Animal models of Parkinson's disease: limits and relevance to neuroprotection studies. Mov. Disord. 28:61–70 [Google Scholar]
  44. Luo Y, Roth GS. 44.  2000. The roles of dopamine oxidative stress and dopamine receptor signaling in aging and age-related neurodegeneration. Antioxid. Redox Signal. 2:449–60 [Google Scholar]
  45. Zheng B, Liao Z, Locascio JJ, Lesniak KA, Roderick SS. 45.  et al. 2010. PGC-1α, a potential therapeutic target for early intervention in Parkinson's disease. Sci. Transl. Med. 2:52ra73 [Google Scholar]
  46. Youdim MB, Oh YJ. 46.  2013. Promise of neurorestoration and mitochondrial biogenesis in Parkinson's disease with multi target drugs: an alternative to stem cell therapy. Exp. Neurobiol. 22:167–72 [Google Scholar]
  47. Scarpulla RC. 47.  2011. Metabolic control of mitochondrial biogenesis through the PGC-1 family regulatory network. Biochim. Biophys. Acta 1813:1269–78 [Google Scholar]
  48. Corona JC, Duchen MR. 48.  2015. PPARγ and PGC-1α as therapeutic targets in Parkinson's. Neurochem. Res. 40:308–16 [Google Scholar]
  49. St-Pierre J, Drori S, Uldry M, Silvaggi JM, Rhee J. 49.  et al. 2006. Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators. Cell 127:397–408 [Google Scholar]
  50. Dulovic M, Jovanovic M, Xilouri M, Stefanis L, Harhaji-Trajkovic L. 50.  et al. 2014. The protective role of AMP-activated protein kinase in alpha-synuclein neurotoxicity in vitro. Neurobiol. Dis. 63:1–11 [Google Scholar]
  51. Martin SD, McGee SL. 51.  2014. The role of mitochondria in the aetiology of insulin resistance and type 2 diabetes. Biochim. Biophys. Acta 1840:1303–12 [Google Scholar]
  52. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S. 52.  et al. 2003. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34:267–73 [Google Scholar]
  53. Calabrese V, Cornelius C, Leso V, Trovato-Salinaro A, Ventimiglia B. 53.  et al. 2012. Oxidative stress, glutathione status, sirtuin and cellular stress response in type 2 diabetes. Biochim. Biophys. Acta 1822:729–36 [Google Scholar]
  54. Robbins NM, Swanson RA. 54.  2014. Opposing effects of glucose on stroke and reperfusion injury: acidosis, oxidative stress, and energy metabolism. Stroke 45:1881–86 [Google Scholar]
  55. Ishihara M. 55.  2012. Acute hyperglycemia in patients with acute myocardial infarction. Circ. J. 76:563–71 [Google Scholar]
  56. Melin J, Hellberg O, Fellstrom B. 56.  2003. Hyperglycaemia and renal ischaemia-reperfusion injury. Nephrol. Dial. Transplant. 18:460–62 [Google Scholar]
  57. Scarpulla RC, Vega RB, Kelly DP. 57.  2012. Transcriptional integration of mitochondrial biogenesis. Trends Endocrinol. Metab. 23:459–66 [Google Scholar]
  58. Kukidome D, Nishikawa T, Sonoda K, Imoto K, Fujisawa K. 58.  et al. 2006. Activation of AMP-activated protein kinase reduces hyperglycemia-induced mitochondrial reactive oxygen species production and promotes mitochondrial biogenesis in human umbilical vein endothelial cells. Diabetes 55:120–27 [Google Scholar]
  59. Golubitzky A, Dan P, Weissman S, Link G, Wikstrom JD, Saada A. 59.  2011. Screening for active small molecules in mitochondrial complex I deficient patient's fibroblasts, reveals AICAR as the most beneficial compound. PLOS ONE 6:e26883 [Google Scholar]
  60. Komen JC, Thorburn DR. 60.  2014. Turn up the power—pharmacological activation of mitochondrial biogenesis in mouse models. Br. J. Pharmacol. 171:1818–36 [Google Scholar]
  61. Canto C, Gerhart-Hines Z, Feige JN, Lagouge M, Noriega L. 61.  et al. 2009. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature 458:1056–60 [Google Scholar]
  62. Dugan LL, You YH, Ali SS, Diamond-Stanic M, Miyamoto S. 62.  et al. 2013. AMPK dysregulation promotes diabetes-related reduction of superoxide and mitochondrial function. J. Clin. Investig. 123:4888–99 [Google Scholar]
  63. Csiszar A, Labinskyy N, Pinto JT, Ballabh P, Zhang H. 63.  et al. 2009. Resveratrol induces mitochondrial biogenesis in endothelial cells. Am. J. Physiol. Heart Circ. Physiol. 297:H13–20 [Google Scholar]
  64. Menzies KJ, Singh K, Saleem A, Hood DA. 64.  2013. Sirtuin 1-mediated effects of exercise and resveratrol on mitochondrial biogenesis. J. Biol. Chem. 288:6968–79 [Google Scholar]
  65. Wang B, Sun J, Ma Y, Wu G, Tian Y. 65.  et al. 2014. Resveratrol preserves mitochondrial function, stimulates mitochondrial biogenesis, and attenuates oxidative stress in regulatory T cells of mice fed a high-fat diet. J. Food Sci. 79:H1823–31 [Google Scholar]
  66. Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H, Lerin C. 66.  et al. 2006. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1α. Cell 127:1109–22 [Google Scholar]
  67. Funk JA, Odejinmi S, Schnellmann RG. 67.  2010. SRT1720 induces mitochondrial biogenesis and rescues mitochondrial function after oxidant injury in renal proximal tubule cells. J. Pharmacol. Exp. Ther. 333:593–601 [Google Scholar]
  68. Minor RK, Baur JA, Gomes AP, Ward TM, Csiszar A. 68.  et al. 2011. SRT1720 improves survival and healthspan of obese mice. Sci. Rep. 1:70 [Google Scholar]
  69. Funk JA, Schnellmann RG. 69.  2013. Accelerated recovery of renal mitochondrial and tubule homeostasis with SIRT1/PGC-1α activation following ischemia-reperfusion injury. Toxicol. Appl. Pharmacol. 273:345–54 [Google Scholar]
  70. Wills LP, Trager RE, Beeson GC, Lindsey CC, Peterson YK. 70.  et al. 2012. The β2-adrenoceptor agonist formoterol stimulates mitochondrial biogenesis. J. Pharmacol. Exp. Ther. 342:106–18 [Google Scholar]
  71. Rasbach KA, Funk JA, Jayavelu T, Green PT, Schnellmann RG. 71.  2010. 5-Hydroxytryptamine receptor stimulation of mitochondrial biogenesis. J. Pharmacol. Exp. Ther. 332:632–39 [Google Scholar]
  72. Tedesco L, Valerio A, Cervino C, Cardile A, Pagano C. 72.  et al. 2008. Cannabinoid type 1 receptor blockade promotes mitochondrial biogenesis through endothelial nitric oxide synthase expression in white adipocytes. Diabetes 57:2028–36 [Google Scholar]
  73. Sarma S. 73.  2012. Use of clinically available PPAR agonists for heart failure; do the risks outweigh the potential benefits?. Curr. Mol. Pharmacol. 5:255–63 [Google Scholar]
  74. Sarma S, Ardehali H, Gheorghiade M. 74.  2012. Enhancing the metabolic substrate: PPAR-α agonists in heart failure. Heart Fail. Rev. 17:35–43 [Google Scholar]
  75. Elijah IE, Borsheim E, Maybauer DM, Finnerty CC, Herndon DN, Maybauer MO. 75.  2012. Role of the PPAR-α agonist fenofibrate in severe pediatric burn. Burns 38:481–86 [Google Scholar]
  76. Cho YR, Lim JH, Kim MY, Kim TW, Hong BY. 76.  et al. 2014. Therapeutic effects of fenofibrate on diabetic peripheral neuropathy by improving endothelial and neural survival in db/db mice. PLOS ONE 9:e83204 [Google Scholar]
  77. Grabacka M, Pierzchalska M, Reiss K. 77.  2013. Peroxisome proliferator activated receptor α ligands as anticancer drugs targeting mitochondrial metabolism. Curr. Pharm. Biotechnol. 14:342–56 [Google Scholar]
  78. Bogacka I, Xie H, Bray GA, Smith SR. 78.  2005. Pioglitazone induces mitochondrial biogenesis in human subcutaneous adipose tissue in vivo. Diabetes 54:1392–99 [Google Scholar]
  79. Pardo R, Enguix N, Lasheras J, Feliu JE, Kralli A, Villena JA. 79.  2011. Rosiglitazone-induced mitochondrial biogenesis in white adipose tissue is independent of peroxisome proliferator-activated receptor-γ coactivator-1α. PLOS ONE 6:e26989 [Google Scholar]
  80. Kleiner S, Nguyen-Tran V, Bare O, Huang X, Spiegelman B, Wu Z. 80.  2009. PPAR{δ} agonism activates fatty acid oxidation via PGC-1{α} but does not increase mitochondrial gene expression and function. J. Biol. Chem. 284:18624–33 [Google Scholar]
  81. Weber K, Bruck P, Mikes Z, Kupper JH, Klingenspor M, Wiesner RJ. 81.  2002. Glucocorticoid hormone stimulates mitochondrial biogenesis specifically in skeletal muscle. Endocrinology 143:177–84 [Google Scholar]
  82. Tiao MM, Lin TK, Chen JB, Liou CW, Wang PW. 82.  et al. 2011. Dexamethasone decreases cholestatic liver injury via inhibition of intrinsic pathway with simultaneous enhancement of mitochondrial biogenesis. Steroids 76:660–66 [Google Scholar]
  83. De Toni L, Strapazzon G, Gianesello L, Caretta N, Pilon C. 83.  et al. 2011. Effects of type 5-phosphodiesterase inhibition on energy metabolism and mitochondrial biogenesis in human adipose tissue ex vivo. J. Endocrinol. Investig. 34:738–41 [Google Scholar]
  84. Nisoli E, Falcone S, Tonello C, Cozzi V, Palomba L. 84.  et al. 2004. Mitochondrial biogenesis by NO yields functionally active mitochondria in mammals. PNAS 101:16507–12 [Google Scholar]
  85. Valerio A, Cardile A, Cozzi V, Bracale R, Tedesco L. 85.  et al. 2006. TNF-α downregulates eNOS expression and mitochondrial biogenesis in fat and muscle of obese rodents. J. Clin. Investig. 116:2791–98 [Google Scholar]
  86. Trevellin E, Scorzeto M, Olivieri M, Granzotto M, Valerio A. 86.  et al. 2014. Exercise training induces mitochondrial biogenesis and glucose uptake in subcutaneous adipose tissue through eNOS-dependent mechanisms. Diabetes 63:2800–11 [Google Scholar]
  87. Vettor R, Valerio A, Ragni M, Trevellin E, Granzotto M. 87.  et al. 2014. Exercise training boosts eNOS-dependent mitochondrial biogenesis in mouse heart: role in adaptation of glucose metabolism. Am. J. Physiol. Endocrinol. Metab. 306:E519–28 [Google Scholar]
  88. Salloum FN, Das A, Samidurai A, Hoke NN, Chau VQ. 88.  et al. 2012. Cinaciguat, a novel activator of soluble guanylate cyclase, protects against ischemia/reperfusion injury: role of hydrogen sulfide. Am. J. Physiol. Heart Circ. Physiol. 302:H1347–54 [Google Scholar]
  89. Vandendriessche B, Rogge E, Goossens V, Vandenabeele P, Stasch JP. 89.  et al. 2013. The soluble guanylate cyclase activator BAY 58-2667 protects against morbidity and mortality in endotoxic shock by recoupling organ systems. PLOS ONE 8:e72155 [Google Scholar]
  90. Miyashita K, Itoh H, Tsujimoto H, Tamura N, Fukunaga Y. 90.  et al. 2009. Natriuretic peptides/cGMP/cGMP-dependent protein kinase cascades promote muscle mitochondrial biogenesis and prevent obesity. Diabetes 58:2880–92 [Google Scholar]
  91. Engeli S, Birkenfeld AL, Badin PM, Bourlier V, Louche K. 91.  et al. 2012. Natriuretic peptides enhance the oxidative capacity of human skeletal muscle. J. Clin. Investig. 122:4675–79 [Google Scholar]
  92. Bordicchia M, Liu D, Amri EZ, Ailhaud G, Dessi-Fulgheri P. 92.  et al. 2012. Cardiac natriuretic peptides act via p38 MAPK to induce the brown fat thermogenic program in mouse and human adipocytes. J. Clin. Investig. 122:1022–36 [Google Scholar]
  93. Rehman H, Krishnasamy Y, Haque K, Thurman RG, Lemasters JJ. 93.  et al. 2014. Green tea polyphenols stimulate mitochondrial biogenesis and improve renal function after chronic cyclosporin a treatment in rats. PLOS ONE 8:e65029 [Google Scholar]
  94. Shen W, Hao J, Feng Z, Tian C, Chen W. 94.  et al. 2011. Lipoamide or lipoic acid stimulates mitochondrial biogenesis in 3T3-L1 adipocytes via the endothelial NO synthase-cGMP-protein kinase G signalling pathway. Br. J. Pharmacol. 162:1213–24 [Google Scholar]
  95. Rasbach KA, Schnellmann RG. 95.  2008. Isoflavones promote mitochondrial biogenesis. J. Pharmacol. Exp. Ther. 325:536–43 [Google Scholar]
  96. Davis JM, Murphy EA, Carmichael MD, Davis B. 96.  2009. Quercetin increases brain and muscle mitochondrial biogenesis and exercise tolerance. Am. J. Physiol. Regul. Integr. Comp. Physiol. 296:R1071–77 [Google Scholar]
  97. Rayamajhi N, Kim SK, Go H, Joe Y, Callaway Z. 97.  et al. 2013. Quercetin induces mitochondrial biogenesis through activation of HO-1 in HepG2 cells. Oxid. Med. Cell Longev. 2013:154279 [Google Scholar]
  98. Thomas RR, Khan SM, Portell FR, Smigrodzki RM, Bennett JP Jr. 98.  2011. Recombinant human mitochondrial transcription factor A stimulates mitochondrial biogenesis and ATP synthesis, improves motor function after MPTP, reduces oxidative stress and increases survival after endotoxin. Mitochondrion 11:108–18 [Google Scholar]
  99. Thomas RR, Khan SM, Smigrodzki RM, Onyango IG, Dennis J. 99.  et al. 2012. RhTFAM treatment stimulates mitochondrial oxidative metabolism and improves memory in aged mice. Aging 4:620–35 [Google Scholar]
  100. Iyer S, Bergquist K, Young K, Gnaiger E, Rao RR, Bennett JP Jr. 100.  2012. Mitochondrial gene therapy improves respiration, biogenesis, and transcription in G11778A Leber's hereditary optic neuropathy and T8993G Leigh's syndrome cells. Hum. Gene Ther. 23:647–57 [Google Scholar]
  101. Hardie DG. 101.  2011. AMP-activated protein kinase: an energy sensor that regulates all aspects of cell function. Genes Dev. 25:1895–908 [Google Scholar]
  102. Narkar VA, Downes M, Yu RT, Embler E, Wang YX. 102.  et al. 2008. AMPK and PPARδ agonists are exercise mimetics. Cell 134:405–15 [Google Scholar]
  103. Russell RR 3rd, Bergeron R, Shulman GI, Young LH. 103.  1999. Translocation of myocardial GLUT-4 and increased glucose uptake through activation of AMPK by AICAR. Am. J. Physiol. 277:H643–49 [Google Scholar]
  104. Lempiainen J, Finckenberg P, Levijoki J, Mervaala E. 104.  2012. AMPK activator AICAR ameliorates ischaemia reperfusion injury in the rat kidney. Br. J. Pharmacol. 166:1905–15 [Google Scholar]
  105. Pold R, Jensen LS, Jessen N, Buhl ES, Schmitz O. 105.  et al. 2005. Long-term AICAR administration and exercise prevents diabetes in ZDF rats. Diabetes 54:928–34 [Google Scholar]
  106. Morigi M, Perico L, Rota C, Longaretti L, Conti S. 106.  et al. 2015. Sirtuin 3-dependent mitochondrial dynamic improvements protect against acute kidney injury. J. Clin. Investig. 125:715–26 [Google Scholar]
  107. Canto C, Auwerx J. 107.  2009. PGC-1α, SIRT1 and AMPK, an energy sensing network that controls energy expenditure. Curr. Opin. Lipidol. 20:98–105 [Google Scholar]
  108. Sun AY, Wang Q, Simonyi A, Sun GY. 108.  2010. Resveratrol as a therapeutic agent for neurodegenerative diseases. Mol. Neurobiol. 41:375–83 [Google Scholar]
  109. Ferretta A, Gaballo A, Tanzarella P, Piccoli C, Capitanio N. 109.  et al. 2014. Effect of resveratrol on mitochondrial function: implications in parkin-associated familiar Parkinson's disease. Biochim. Biophys. Acta 1842:902–15 [Google Scholar]
  110. Pasinetti GM, Wang J, Marambaud P, Ferruzzi M, Gregor P. 110.  et al. 2011. Neuroprotective and metabolic effects of resveratrol: therapeutic implications for Huntington's disease and other neurodegenerative disorders. Exp. Neurol. 232:1–6 [Google Scholar]
  111. Petrovski G, Gurusamy N, Das DK. 111.  2011. Resveratrol in cardiovascular health and disease. Ann. N.Y. Acad. Sci. 1215:22–33 [Google Scholar]
  112. Kanamori H, Takemura G, Goto K, Tsujimoto A, Ogino A. 112.  et al. 2013. Resveratrol reverses remodeling in hearts with large, old myocardial infarctions through enhanced autophagy-activating AMP kinase pathway. Am. J. Pathol. 182:701–13 [Google Scholar]
  113. Rivera L, Moron R, Zarzuelo A, Galisteo M. 113.  2009. Long-term resveratrol administration reduces metabolic disturbances and lowers blood pressure in obese Zucker rats. Biochem. Pharmacol. 77:1053–63 [Google Scholar]
  114. Beaudeux JL, Nivet-Antoine V, Giral P. 114.  2010. Resveratrol: a relevant pharmacological approach for the treatment of metabolic syndrome?. Curr. Opin. Clin. Nutr. Metab. Care 13:729–36 [Google Scholar]
  115. Brasnyo P, Molnar GA, Mohas M, Marko L, Laczy B. 115.  et al. 2011. Resveratrol improves insulin sensitivity, reduces oxidative stress and activates the Akt pathway in type 2 diabetic patients. Br. J. Nutr. 106:383–89 [Google Scholar]
  116. Milne JC, Lambert PD, Schenk S, Carney DP, Smith JJ. 116.  et al. 2007. Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature 450:712–16 [Google Scholar]
  117. de Boer VC, de Goffau MC, Arts IC, Hollman PC, Keijer J. 117.  2006. SIRT1 stimulation by polyphenols is affected by their stability and metabolism. Mech. Ageing Dev. 127:618–27 [Google Scholar]
  118. Haigis MC, Deng CX, Finley LW, Kim HS, Gius D. 118.  2012. SIRT3 is a mitochondrial tumor suppressor: a scientific tale that connects aberrant cellular ROS, the Warburg effect, and carcinogenesis. Cancer Res. 72:2468–72 [Google Scholar]
  119. Puigserver P, Spiegelman BM. 119.  2003. Peroxisome proliferator-activated receptor-γ coactivator 1 α (PGC-1α): transcriptional coactivator and metabolic regulator. Endocr. Rev. 24:78–90 [Google Scholar]
  120. Hoshino D, Yoshida Y, Holloway GP, Lally J, Hatta H, Bonen A. 120.  2012. Clenbuterol, a β2-adrenergic agonist, reciprocally alters PGC-1α and RIP140 and reduces fatty acid and pyruvate oxidation in rat skeletal muscle. Am. J. Physiol. Regul. Integr. Comp. Physiol. 302:R373–84 [Google Scholar]
  121. Peterson YK, Cameron RB, Wills LP, Trager RE, Lindsey CC. 121.  et al. 2013. β2-adrenoceptor agonists in the regulation of mitochondrial biogenesis. Bioorg. Med. Chem. Lett. 23:5376–81 [Google Scholar]
  122. Nisoli E, Tonello C, Cardile A, Cozzi V, Bracale R. 122.  et al. 2005. Calorie restriction promotes mitochondrial biogenesis by inducing the expression of eNOS. Science 310:314–17 [Google Scholar]
  123. Nisoli E, Clementi E, Paolucci C, Cozzi V, Tonello C. 123.  et al. 2003. Mitochondrial biogenesis in mammals: the role of endogenous nitric oxide. Science 299:896–99 [Google Scholar]
  124. Nisoli E, Carruba MO. 124.  2006. Nitric oxide and mitochondrial biogenesis. J. Cell Sci. 119:2855–62 [Google Scholar]
  125. Miglio G, Rosa AC, Rattazzi L, Collino M, Lombardi G, Fantozzi R. 125.  2009. PPARγ stimulation promotes mitochondrial biogenesis and prevents glucose deprivation-induced neuronal cell loss. Neurochem. Int. 55:496–504 [Google Scholar]
  126. Wu QQ, Wang Y, Senitko M, Meyer C, Wigley WC. 126.  et al. 2011. Bardoxolone methyl (BARD) ameliorates ischemic AKI and increases expression of protective genes Nrf2, PPARγ, and HO-1. Am. J. Physiol. Ren. Physiol. 300:F1180–92 [Google Scholar]
  127. Singh JP, Singh AP, Bhatti R. 127.  2014. Explicit role of peroxisome proliferator-activated receptor-γ in gallic acid-mediated protection against ischemia-reperfusion-induced acute kidney injury in rats. J. Surg. Res. 187:631–39 [Google Scholar]
  128. Funk JA, Schnellmann RG. 128.  2012. Persistent disruption of mitochondrial homeostasis after acute kidney injury. Am. J. Physiol. Ren. Physiol. 302:F853–64 [Google Scholar]
  129. Stallons LJ, Whitaker RM, Schnellmann RG. 129.  2014. Suppressed mitochondrial biogenesis in folic acid-induced acute kidney injury and early fibrosis. Toxicol. Lett. 224:326–32 [Google Scholar]
  130. Smith JA, Stallons LJ, Collier JB, Chavin KD, Schnellmann RG. 130.  2015. Suppression of mitochondrial biogenesis through toll-like receptor 4-dependent mitogen-activated protein kinase kinase/extracellular signal-regulated kinase signaling in endotoxin-induced acute kidney injury. J. Pharmacol. Exp. Ther. 352:346–57 [Google Scholar]
  131. Rong JX, Qiu Y, Hansen MK, Zhu L, Zhang V. 131.  et al. 2007. Adipose mitochondrial biogenesis is suppressed in db/db and high-fat diet-fed mice and improved by rosiglitazone. Diabetes 56:1751–60 [Google Scholar]
  132. Chistiakov DA, Sobenin IA, Revin VV, Orekhov AN, Bobryshev YV. 132.  2014. Mitochondrial aging and age-related dysfunction of mitochondria. Biomed. Res. Int. 2014:238463 [Google Scholar]
  133. Koltai E, Hart N, Taylor AW, Goto S, Ngo JK. 133.  et al. 2012. Age-associated declines in mitochondrial biogenesis and protein quality control factors are minimized by exercise training. Am. J. Physiol. Regul. Integr. Comp. Physiol. 303:R127–34 [Google Scholar]
  134. Palomer X, Alvarez-Guardia D, Rodriguez-Calvo R, Coll T, Laguna JC. 134.  et al. 2009. TNF-α reduces PGC-1α expression through NF-κB and p38 MAPK leading to increased glucose oxidation in a human cardiac cell model. Cardiovasc. Res. 81:703–12 [Google Scholar]
  135. Piantadosi CA, Withers CM, Bartz RR, MacGarvey NC, Fu P. 135.  et al. 2011. Heme oxygenase-1 couples activation of mitochondrial biogenesis to anti-inflammatory cytokine expression. J. Biol. Chem. 286:16374–85 [Google Scholar]
  136. Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T. 136.  et al. 2010. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature 464:104–7 [Google Scholar]
  137. Ye L, Kleiner S, Wu J, Sah R, Gupta RK. 137.  et al. 2012. TRPV4 is a regulator of adipose oxidative metabolism, inflammation, and energy homeostasis. Cell 151:96–110 [Google Scholar]
  138. Arany Z, Wagner BK, Ma Y, Chinsomboon J, Laznik D, Spiegelman BM. 138.  2008. Gene expression-based screening identifies microtubule inhibitors as inducers of PGC-1α and oxidative phosphorylation. PNAS 105:4721–26 [Google Scholar]
  139. Beeson CC, Beeson GC, Schnellmann RG. 139.  2010. A high-throughput respirometric assay for mitochondrial biogenesis and toxicity. Anal. Biochem. 404:75–81 [Google Scholar]
  140. Wills LP, Beeson GC, Trager RE, Lindsey CC, Beeson CC. 140.  et al. 2013. High-throughput respirometric assay identifies predictive toxicophore of mitochondrial injury. Toxicol. Appl. Pharmacol. 272:490–502 [Google Scholar]
  141. Ong SB, Subrayan S, Lim SY, Yellon DM, Davidson SM, Hausenloy DJ. 141.  2010. Inhibiting mitochondrial fission protects the heart against ischemia/reperfusion injury. Circulation 121:2012–22 [Google Scholar]
  142. Chen KH, Guo X, Ma D, Guo Y, Li Q. 142.  et al. 2004. Dysregulation of HSG triggers vascular proliferative disorders. Nat. Cell Biol. 6:872–83 [Google Scholar]
  143. Liu T, Xue CC, Shi YL, Bai XJ, Li ZF, Yi CL. 143.  2014. Overexpression of mitofusin 2 inhibits reactive astrogliosis proliferation in vitro. Neurosci. Lett. 579:24–29 [Google Scholar]
  144. Yanagishita T, Tomita M, Itoh S, Mukae S, Arata H. 144.  et al. 1997. Protective effect of captopril on ischemic myocardium. Jpn. Circ. J. 61:161–69 [Google Scholar]
  145. Monaco EA 3rd, Weiner GM, Friedlander RM. 145.  2013. Randomized-controlled trial of minocycline for spinal cord injury shows promise. Neurosurgery 72:N17–19 [Google Scholar]
/content/journals/10.1146/annurev-pharmtox-010715-103155
Loading
/content/journals/10.1146/annurev-pharmtox-010715-103155
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