Skip to main content

Advertisement

Log in

Energy dysfunction in Huntington’s disease: insights from PGC-1α, AMPK, and CKB

  • Review
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

Huntington’s disease (HD) is an autosomal dominant neurodegenerative disease caused by a CAG trinucleotide expansion in the Huntingtin (Htt) gene. When the number of CAG repeats exceeds 36, the translated polyglutamine-expanded Htt protein interferes with the normal functions of many types of cellular machinery and causes cytotoxicity. Clinical symptoms include progressive involuntary movement disorders, psychiatric signs, cognitive decline, dementia, and a shortened lifespan. The most severe brain atrophy is observed in the striatum and cortex. Besides the well-characterized neuronal defects, recent studies showed that the functions of mitochondria and several key players in energy homeostasis are abnormally regulated during HD progression. Energy dysregulation thus is now recognized as an important pathogenic pathway of HD. This review focuses on the importance of three key molecular determinants (peroxisome proliferator-activated receptor-γ coactivator-1α, AMP-activated protein kinase, and creatine kinase B) of cellular energy homeostasis and their possible involvement in HD pathogenesis.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Martin JB, Gusella JF (1986) Huntington’s disease. Pathogenesis and management. N Engl J Med 315:1267–1276

    Article  PubMed  CAS  Google Scholar 

  2. The Huntington’s Disease Collaborative Research Group (1993) A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. Cell 72:971–983

    Article  Google Scholar 

  3. Landles C, Bates GP (2004) Huntingtin and the molecular pathogenesis of Huntington’s disease. Fourth in molecular medicine review series. EMBO Rep 5:958–963

    Article  PubMed  CAS  Google Scholar 

  4. Buckley NJ, Johnson R, Zuccato C, Bithell A, Cattaneo E (2010) The role of REST in transcriptional and epigenetic dysregulation in Huntington’s disease. Neurobiol Dis 39:28–39

    Google Scholar 

  5. Li H, Li SH, Yu ZX, Shelbourne P, Li XJ (2001) Huntingtin aggregate-associated axonal degeneration is an early pathological event in Huntington’s disease mice. J Neurosci 21:8473–8481

    PubMed  CAS  Google Scholar 

  6. Lin YS, Chen CM, Soong BW, Wu YR, Chen HM, Yeh WY, Wu DR, Lin YJ, Poon PW, Cheng ML, Wang CH, Chern Y (2011) Dysregulated brain creatine kinase is associated with hearing impairment in mouse models of Huntington disease. J Clin Invest 121:1519–1523

    Article  PubMed  CAS  Google Scholar 

  7. Chiang MC, Chen HM, Lee YH, Chang HH, Wu YC, Soong BW, Chen CM, Wu YR, Liu CS, Niu DM, Wu JY, Chen YT, Chern Y (2007) Dysregulation of C/EBPalpha by mutant Huntingtin causes the urea cycle deficiency in Huntington’s disease. Hum Mol Genet 16:483–498

    Article  PubMed  CAS  Google Scholar 

  8. Klapstein GJ, Fisher RS, Zanjani H, Cepeda C, Jokel ES, Chesselet MF, Levine MS (2001) Electrophysiological and morphological changes in striatal spiny neurons in R6/2 Huntington’s disease transgenic mice. J Neurophysiol 86:2667–2677

    PubMed  CAS  Google Scholar 

  9. Martindale D, Hackam A, Wieczorek A, Ellerby L, Wellington C, McCutcheon K, Singaraja R, Kazemi-Esfarjani P, Devon R, Kim SU, Bredesen DE, Tufaro F, Hayden MR (1998) Length of huntingtin and its polyglutamine tract influences localization and frequency of intracellular aggregates. Nat Genet 18:150–154

    Article  PubMed  CAS  Google Scholar 

  10. Vonsattel JP, Myers RH, Stevens TJ, Ferrante RJ, Bird ED, Richardson EP Jr (1985) Neuropathological classification of Huntington’s disease. J Neuropathol Exp Neurol 44:559–577

    Article  PubMed  CAS  Google Scholar 

  11. Borrell-Pages M, Zala D, Humbert S, Saudou F (2006) Huntington’s disease: from huntingtin function and dysfunction to therapeutic strategies. Cell Mol Life Sci 63:2642–2660

    Article  PubMed  CAS  Google Scholar 

  12. Oliveira JM (2010) Nature and cause of mitochondrial dysfunction in Huntington’s disease: focusing on huntingtin and the striatum. J Neurochem 114:1–12

    PubMed  CAS  Google Scholar 

  13. Subramaniam S, Snyder SH (2011) Huntington’s disease is a disorder of the corpus striatum: focus on Rhes (Ras homologue enriched in the striatum). Neuropharmacology 60:1187–1192

    Article  PubMed  CAS  Google Scholar 

  14. Khoshnan A, Patterson PH (2011) The role of IkappaB kinase complex in the neurobiology of Huntington’s disease. Neurobiol Dis 43:305–311

    Article  PubMed  CAS  Google Scholar 

  15. Crook ZR, Housman D (2011) Huntington’s disease: can mice lead the way to treatment? Neuron 69:423–435

    Article  PubMed  CAS  Google Scholar 

  16. Ross CA, Tabrizi SJ (2011) Huntington’s disease: from molecular pathogenesis to clinical treatment. Lancet Neurol 10:83–98

    Article  PubMed  CAS  Google Scholar 

  17. Pratley RE, Salbe AD, Ravussin E, Caviness JN (2000) Higher sedentary energy expenditure in patients with Huntington’s disease. Ann Neurol 47:64–70

    Article  PubMed  CAS  Google Scholar 

  18. Hurlbert MS, Zhou W, Wasmeier C, Kaddis FG, Hutton JC, Freed CR (1999) Mice transgenic for an expanded CAG repeat in the Huntington’s disease gene develop diabetes. Diabetes 48:649–651

    Article  PubMed  CAS  Google Scholar 

  19. Josefsen K, Nielsen MD, Jorgensen KH, Bock T, Norremolle A, Sorensen SA, Naver B, Hasholt L (2008) Impaired glucose tolerance in the R6/1 transgenic mouse model of Huntington’s disease. J Neuroendocrinol 20:165–172

    Article  PubMed  CAS  Google Scholar 

  20. Trushina E, Singh RD, Dyer RB, Cao S, Shah VH, Parton RG, Pagano RE, McMurray CT (2006) Mutant huntingtin inhibits clathrin-independent endocytosis and causes accumulation of cholesterol in vitro and in vivo. Hum Mol Genet 15:3578–3591

    Article  PubMed  CAS  Google Scholar 

  21. Valenza M, Rigamonti D, Goffredo D, Zuccato C, Fenu S, Jamot L, Strand A, Tarditi A, Woodman B, Racchi M, Mariotti C, Di Donato S, Corsini A, Bates G, Pruss R, Olson JM, Sipione S, Tartari M, Cattaneo E (2005) Dysfunction of the cholesterol biosynthetic pathway in Huntington’s disease. J Neurosci 25:9932–9939

    Article  PubMed  CAS  Google Scholar 

  22. Valenza M, Carroll JB, Leoni V, Bertram LN, Bjorkhem I, Singaraja RR, Di Donato S, Lutjohann D, Hayden MR, Cattaneo E (2007) Cholesterol biosynthesis pathway is disturbed in YAC128 mice and is modulated by huntingtin mutation. Hum Mol Genet 16:2187–2198

    Article  PubMed  CAS  Google Scholar 

  23. Morton AJ, Lagan MA, Skepper JN, Dunnett SB (2000) Progressive formation of inclusions in the striatum and hippocampus of mice transgenic for the human Huntington’s disease mutation. J Neurocytol 29:679–702

    Article  PubMed  CAS  Google Scholar 

  24. Karasinska JM, Hayden MR (2011) Cholesterol metabolism in Huntington disease. Nat Rev Neurol 7:561–572

    Article  PubMed  CAS  Google Scholar 

  25. Chiang MC, Chen HM, Lai HL, Chen HW, Chou SY, Chen CM, Tsai FJ, Chern Y (2009) The A2A adenosine receptor rescues the urea cycle deficiency of Huntington’s disease by enhancing the activity of the ubiquitin–proteasome system. Hum Mol Genet 18:2929–2942

    Article  PubMed  CAS  Google Scholar 

  26. Podolsky S, Leopold NA, Sax DS (1972) Increased frequency of diabetes mellitus in patients with Huntington’s chorea. Lancet 1:1356–1358

    Article  PubMed  CAS  Google Scholar 

  27. Squitieri F, Cannella M, Sgarbi G, Maglione V, Falleni A, Lenzi P, Baracca A, Cislaghi G, Saft C, Ragona G, Russo MA, Thompson LM, Solaini G, Fornai F (2006) Severe ultrastructural mitochondrial changes in lymphoblasts homozygous for Huntington disease mutation. Mech Ageing Dev 127:217–220

    Article  PubMed  CAS  Google Scholar 

  28. Orr AL, Li S, Wang CE, Li H, Wang J, Rong J, Xu X, Mastroberardino PG, Greenamyre JT, Li XJ (2008) N-terminal mutant huntingtin associates with mitochondria and impairs mitochondrial trafficking. J Neurosci 28:2783–2792

    Article  PubMed  CAS  Google Scholar 

  29. Almeida S, Sarmento-Ribeiro AB, Januario C, Rego AC, Oliveira CR (2008) Evidence of apoptosis and mitochondrial abnormalities in peripheral blood cells of Huntington’s disease patients. Biochem Biophys Res Commun 374:599–603

    Article  PubMed  CAS  Google Scholar 

  30. Panov AV, Gutekunst CA, Leavitt BR, Hayden MR, Burke JR, Strittmatter WJ, Greenamyre JT (2002) Early mitochondrial calcium defects in Huntington’s disease are a direct effect of polyglutamines. Nat Neurosci 5:731–736

    PubMed  CAS  Google Scholar 

  31. Panov AV, Burke JR, Strittmatter WJ, Greenamyre JT (2003) In vitro effects of polyglutamine tracts on Ca2+-dependent depolarization of rat and human mitochondria: relevance to Huntington’s disease. Arch Biochem Biophys 410:1–6

    Article  PubMed  CAS  Google Scholar 

  32. Milakovic T, Quintanilla RA, Johnson GV (2006) Mutant huntingtin expression induces mitochondrial calcium handling defects in clonal striatal cells: functional consequences. J Biol Chem 281:34785–34795

    Article  PubMed  CAS  Google Scholar 

  33. Song W, Chen J, Petrilli A, Liot G, Klinglmayr E, Zhou Y, Poquiz P, Tjong J, Pouladi MA, Hayden MR, Masliah E, Ellisman M, Rouiller I, Schwarzenbacher R, Bossy B, Perkins G, Bossy-Wetzel E (2011) Mutant huntingtin binds the mitochondrial fission GTPase dynamin-related protein-1 and increases its enzymatic activity. Nat Med 17:377–382

    Article  PubMed  CAS  Google Scholar 

  34. Wang H, Lim PJ, Karbowski M, Monteiro MJ (2009) Effects of overexpression of huntingtin proteins on mitochondrial integrity. Hum Mol Genet 18:737–752

    Article  PubMed  CAS  Google Scholar 

  35. Browne SE, Bowling AC, MacGarvey U, Baik MJ, Berger SC, Muqit MM, Bird ED, Beal MF (1997) Oxidative damage and metabolic dysfunction in Huntington’s disease: selective vulnerability of the basal ganglia. Ann Neurol 41:646–653

    Article  PubMed  CAS  Google Scholar 

  36. Gu M, Gash MT, Mann VM, Javoy-Agid F, Cooper JM, Schapira AH (1996) Mitochondrial defect in Huntington’s disease caudate nucleus. Ann Neurol 39:385–389

    Article  PubMed  CAS  Google Scholar 

  37. Benchoua A, Trioulier Y, Zala D, Gaillard MC, Lefort N, Dufour N, Saudou F, Elalouf JM, Hirsch E, Hantraye P, Deglon N, Brouillet E (2006) Involvement of mitochondrial complex II defects in neuronal death produced by N-terminus fragment of mutated huntingtin. Mol Biol Cell 17:1652–1663

    Article  PubMed  CAS  Google Scholar 

  38. Ferrante RJ, Andreassen OA, Dedeoglu A, Ferrante KL, Jenkins BG, Hersch SM, Beal MF (2002) Therapeutic effects of coenzyme Q10 and remacemide in transgenic mouse models of Huntington’s disease. J Neurosci 22:1592–1599

    PubMed  CAS  Google Scholar 

  39. Fontaine MA, Geddes JW, Banks A, Butterfield DA (2000) Effect of exogenous and endogenous antioxidants on 3-nitropionic acid-induced in vivo oxidative stress and striatal lesions: insights into Huntington’s disease. J Neurochem 75:1709–1715

    Article  PubMed  CAS  Google Scholar 

  40. Stack EC, Matson WR, Ferrante RJ (2008) Evidence of oxidant damage in Huntington’s disease: translational strategies using antioxidants. Ann N Y Acad Sci 1147:79–92

    Article  PubMed  CAS  Google Scholar 

  41. Andreassen OA, Ferrante RJ, Dedeoglu A, Beal MF (2001) Lipoic acid improves survival in transgenic mouse models of Huntington’s disease. Neuroreport 12:3371–3373

    Article  PubMed  CAS  Google Scholar 

  42. Keene CD, Rodrigues CM, Eich T, Chhabra MS, Steer CJ, Low WC (2002) Tauroursodeoxycholic acid, a bile acid, is neuroprotective in a transgenic animal model of Huntington’s disease. Proc Natl Acad Sci USA 99:10671–10676

    Article  PubMed  CAS  Google Scholar 

  43. Fernandes HB, Baimbridge KG, Church J, Hayden MR, Raymond LA (2007) Mitochondrial sensitivity and altered calcium handling underlie enhanced NMDA-induced apoptosis in YAC128 model of Huntington’s disease. J Neurosci 27:13614–13623

    Article  PubMed  CAS  Google Scholar 

  44. Ju TC, Chen HM, Lin JT, Chang CP, Chang WC, Kang JJ, Sun CP, Tao MH, Tu PH, Chang C, Dickson DW, Chern Y (2011) Nuclear translocation of AMPK-alpha1 potentiates striatal neurodegeneration in Huntington’s disease. J Cell Biol 194:209–227

    Article  PubMed  CAS  Google Scholar 

  45. Bossy-Wetzel E, Petrilli A, Knott AB (2008) Mutant huntingtin and mitochondrial dysfunction. Trends Neurosci 31:609–616

    Article  PubMed  CAS  Google Scholar 

  46. Li XJ, Orr AL, Li S (2010) Impaired mitochondrial trafficking in Huntington’s disease. Biochim Biophys Acta 1802:62–65

    Article  PubMed  CAS  Google Scholar 

  47. Chang DT, Rintoul GL, Pandipati S, Reynolds IJ (2006) Mutant huntingtin aggregates impair mitochondrial movement and trafficking in cortical neurons. Neurobiol Dis 22:388–400

    Article  PubMed  CAS  Google Scholar 

  48. Naya FJ, Mercer B, Shelton J, Richardson JA, Williams RS, Olson EN (2000) Stimulation of slow skeletal muscle fiber gene expression by calcineurin in vivo. J Biol Chem 275:4545–4548

    Article  PubMed  CAS  Google Scholar 

  49. Huss JM, Torra IP, Staels B, Giguere V, Kelly DP (2004) Estrogen-related receptor alpha directs peroxisome proliferator-activated receptor alpha signaling in the transcriptional control of energy metabolism in cardiac and skeletal muscle. Mol Cell Biol 24:9079–9091

    Article  PubMed  CAS  Google Scholar 

  50. Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Mootha V, Troy A, Cinti S, Lowell B, Scarpulla RC, Spiegelman BM (1999) Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1. Cell 98:115–124

    Article  PubMed  CAS  Google Scholar 

  51. Puigserver P, Wu Z, Park CW, Graves R, Wright M, Spiegelman BM (1998) A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell 92:829–839

    Article  PubMed  CAS  Google Scholar 

  52. Huss JM, Kopp RP, Kelly DP (2002) Peroxisome proliferator-activated receptor coactivator-1alpha (PGC-1alpha) coactivates the cardiac-enriched nuclear receptors estrogen-related receptor-alpha and -gamma. Identification of novel leucine-rich interaction motif within PGC-1alpha. J Biol Chem 277:40265–40274

    Article  PubMed  CAS  Google Scholar 

  53. Zhang Y, Castellani LW, Sinal CJ, Gonzalez FJ, Edwards PA (2004) Peroxisome proliferator-activated receptor-gamma coactivator 1alpha (PGC-1alpha) regulates triglyceride metabolism by activation of the nuclear receptor FXR. Genes Dev 18:157–169

    Article  PubMed  CAS  Google Scholar 

  54. Desvergne B, Wahli W (1999) Peroxisome proliferator-activated receptors: nuclear control of metabolism. Endocr Rev 20:649–688

    Article  PubMed  CAS  Google Scholar 

  55. Southgate RJ, Bruce CR, Carey AL, Steinberg GR, Walder K, Monks R, Watt MJ, Hawley JA, Birnbaum MJ, Febbraio MA (2005) PGC-1alpha gene expression is down-regulated by Akt-mediated phosphorylation and nuclear exclusion of FoxO1 in insulin-stimulated skeletal muscle. FASEB J 19:2072–2074

    PubMed  CAS  Google Scholar 

  56. McGee SL, Hargreaves M (2004) Exercise and myocyte enhancer factor 2 regulation in human skeletal muscle. Diabetes 53:1208–1214

    Article  PubMed  CAS  Google Scholar 

  57. Oberkofler H, Schraml E, Krempler F, Patsch W (2004) Restoration of sterol-regulatory-element-binding protein-1c gene expression in HepG2 cells by peroxisome–proliferator-activated receptor-gamma co-activator-1alpha. Biochem J 381:357–363

    Article  PubMed  CAS  Google Scholar 

  58. Yoon JC, Puigserver P, Chen G, Donovan J, Wu Z, Rhee J, Adelmant G, Stafford J, Kahn CR, Granner DK, Newgard CB, Spiegelman BM (2001) Control of hepatic gluconeogenesis through the transcriptional coactivator PGC-1. Nature 413:131–138

    Article  PubMed  CAS  Google Scholar 

  59. Lelliott C, Vidal-Puig AJ (2004) Lipotoxicity, an imbalance between lipogenesis de novo and fatty acid oxidation. Int J Obes Relat Metab Disord 28(Suppl 4):S22–S28

    Article  PubMed  CAS  Google Scholar 

  60. Koo SH, Satoh H, Herzig S, Lee CH, Hedrick S, Kulkarni R, Evans RM, Olefsky J, Montminy M (2004) PGC-1 promotes insulin resistance in liver through PPAR-alpha-dependent induction of TRB-3. Nat Med 10:530–534

    Article  PubMed  CAS  Google Scholar 

  61. Nemoto S, Fergusson MM, Finkel T (2005) SIRT1 functionally interacts with the metabolic regulator and transcriptional coactivator PGC-1{alpha}. J Biol Chem 280:16456–16460

    Article  PubMed  CAS  Google Scholar 

  62. Jager S, Handschin C, St-Pierre J, Spiegelman BM (2007) AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha. Proc Natl Acad Sci USA 104:12017–12022

    Article  PubMed  CAS  Google Scholar 

  63. Puigserver P, Rhee J, Lin J, Wu Z, Yoon JC, Zhang CY, Krauss S, Mootha VK, Lowell BB, Spiegelman BM (2001) Cytokine stimulation of energy expenditure through p38 MAP kinase activation of PPARgamma coactivator-1. Mol Cell 8:971–982

    Article  PubMed  CAS  Google Scholar 

  64. Teyssier C, Ma H, Emter R, Kralli A, Stallcup MR (2005) Activation of nuclear receptor coactivator PGC-1alpha by arginine methylation. Genes Dev 19:1466–1473

    Article  PubMed  CAS  Google Scholar 

  65. Rytinki MM, Palvimo JJ (2009) SUMOylation attenuates the function of PGC-1alpha. J Biol Chem 284:26184–26193

    Article  PubMed  CAS  Google Scholar 

  66. Cui L, Jeong H, Borovecki F, Parkhurst CN, Tanese N, Krainc D (2006) Transcriptional repression of PGC-1alpha by mutant huntingtin leads to mitochondrial dysfunction and neurodegeneration. Cell 127:59–69

    Article  PubMed  CAS  Google Scholar 

  67. Weydt P, Pineda VV, Torrence AE, Libby RT, Satterfield TF, Lazarowski ER, Gilbert ML, Morton GJ, Bammler TK, Strand AD, Cui L, Beyer RP, Easley CN, Smith AC, Krainc D, Luquet S, Sweet IR, Schwartz MW, La Spada AR (2006) Thermoregulatory and metabolic defects in Huntington’s disease transgenic mice implicate PGC-1alpha in Huntington’s disease neurodegeneration. Cell Metab 4:349–362

    Article  PubMed  CAS  Google Scholar 

  68. Chaturvedi RK, Adhihetty P, Shukla S, Hennessy T, Calingasan N, Yang L, Starkov A, Kiaei M, Cannella M, Sassone J, Ciammola A, Squitieri F, Beal MF (2009) Impaired PGC-1alpha function in muscle in Huntington’s disease. Hum Mol Genet 18:3048–3065

    Article  PubMed  CAS  Google Scholar 

  69. Xiang Z, Valenza M, Cui L, Leoni V, Jeong HK, Brilli E, Zhang J, Peng Q, Duan W, Reeves SA, Cattaneo E, Krainc D (2011) Peroxisome-proliferator-activated receptor gamma coactivator 1 alpha contributes to dysmyelination in experimental models of Huntington’s disease. J Neurosci 31:9544–9553

    Article  PubMed  CAS  Google Scholar 

  70. Leone TC, Lehman JJ, Finck BN, Schaeffer PJ, Wende AR, Boudina S, Courtois M, Wozniak DF, Sambandam N, Bernal-Mizrachi C, Chen Z, Holloszy JO, Medeiros DM, Schmidt RE, Saffitz JE, Abel ED, Semenkovich CF, Kelly DP (2005) PGC-1alpha deficiency causes multi-system energy metabolic derangements: muscle dysfunction, abnormal weight control and hepatic steatosis. PLoS Biol 3:e101

    Article  PubMed  CAS  Google Scholar 

  71. Lin J, Wu PH, Tarr PT, Lindenberg KS, St-Pierre J, Zhang CY, Mootha VK, Jager S, Vianna CR, Reznick RM, Cui L, Manieri M, Donovan MX, Wu Z, Cooper MP, Fan MC, Rohas LM, Zavacki AM, Cinti S, Shulman GI, Lowell BB, Krainc D, Spiegelman BM (2004) Defects in adaptive energy metabolism with CNS-linked hyperactivity in PGC-1alpha null mice. Cell 119:121–135

    Article  PubMed  CAS  Google Scholar 

  72. St-Pierre J, Drori S, Uldry M, Silvaggi JM, Rhee J, Jager S, Handschin C, Zheng K, Lin J, Yang W, Simon DK, Bachoo R, Spiegelman BM (2006) Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators. Cell 127:397–408

    Article  PubMed  CAS  Google Scholar 

  73. Chiang MC, Chern Y, Juo CG (2011) The dysfunction of hepatic transcriptional factors in mice with Huntington’s Disease. Biochim Biophys Acta 1812:1111–1120

    Article  PubMed  CAS  Google Scholar 

  74. Chiang MC, Chern Y, Huang RN (2012) PPARgamma rescue of the mitochondrial dysfunction in Huntington’s disease. Neurobiol Dis 45:322–328

    Article  PubMed  CAS  Google Scholar 

  75. Chiang MC, Chen CM, Lee MR, Chen HW, Chen HM, Wu YS, Hung CH, Kang JJ, Chang CP, Chang C, Wu YR, Tsai YS, Chern Y (2010) Modulation of energy deficiency in Huntington’s disease via activation of the peroxisome proliferator-activated receptor gamma. Hum Mol Genet 19:4043–4058

    Article  PubMed  CAS  Google Scholar 

  76. Canto C, Auwerx J (2009) PGC-1alpha, SIRT1 and AMPK, an energy sensing network that controls energy expenditure. Curr Opin Lipidol 20:98–105

    Article  PubMed  CAS  Google Scholar 

  77. Jiang M, Wang J, Fu J, Du L, Jeong H, West T, Xiang L, Peng Q, Hou Z, Cai H, Seredenina T, Arbez N, Zhu S, Sommers K, Qian J, Zhang J, Mori S, Yang XW, Tamashiro KL, Aja S, Moran TH, Luthi-Carter R, Martin B, Maudsley S, Mattson MP, Cichewicz RH, Ross CA, Holtzman DM, Krainc D, Duan W (2012) Neuroprotective role of Sirt1 in mammalian models of Huntington’s disease through activation of multiple Sirt1 targets. Nat Med 18:153–158

    Article  CAS  Google Scholar 

  78. Long YC, Zierath JR (2006) AMP-activated protein kinase signaling in metabolic regulation. J Clin Invest 116:1776–1783

    Article  PubMed  CAS  Google Scholar 

  79. Ramamurthy S, Ronnett GV (2006) Developing a head for energy sensing: AMP-activated protein kinase as a multifunctional metabolic sensor in the brain. J Physiol 574:85–93

    Article  PubMed  CAS  Google Scholar 

  80. Hardie DG (2008) AMPK: a key regulator of energy balance in the single cell and the whole organism. Int J Obes (Lond) 32(Suppl 4):S7–S12

    Article  CAS  Google Scholar 

  81. Lage R, Dieguez C, Vidal-Puig A, Lopez M (2008) AMPK: a metabolic gauge regulating whole-body energy homeostasis. Trends Mol Med 14:539–549

    Article  PubMed  CAS  Google Scholar 

  82. Greer EL, Oskoui PR, Banko MR, Maniar JM, Gygi MP, Gygi SP, Brunet A (2007) The energy sensor AMP-activated protein kinase directly regulates the mammalian FOXO3 transcription factor. J Biol Chem 282:30107–30119

    Article  PubMed  CAS  Google Scholar 

  83. Canto C, Jiang LQ, Deshmukh AS, Mataki C, Coste A, Lagouge M, Zierath JR, Auwerx J (2010) Interdependence of AMPK and SIRT1 for metabolic adaptation to fasting and exercise in skeletal muscle. Cell Metab 11:213–219

    Article  PubMed  CAS  Google Scholar 

  84. Hardie DG, Ross FA, Hawley SA (2012) AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol 13:251–262

    Article  PubMed  CAS  Google Scholar 

  85. Steinberg GR, Kemp BE (2009) AMPK in health and disease. Physiol Rev 89:1025–1078

    Article  PubMed  CAS  Google Scholar 

  86. Kemp BE, Mitchelhill KI, Stapleton D, Michell BJ, Chen ZP, Witters LA (1999) Dealing with energy demand: the AMP-activated protein kinase. Trends Biochem Sci 24:22–25

    Article  PubMed  CAS  Google Scholar 

  87. Salt I, Celler JW, Hawley SA, Prescott A, Woods A, Carling D, Hardie DG (1998) AMP-activated protein kinase: greater AMP dependence, and preferential nuclear localization, of complexes containing the alpha2 isoform. Biochem J 334(Pt 1):177–187

    PubMed  CAS  Google Scholar 

  88. Turnley AM, Stapleton D, Mann RJ, Witters LA, Kemp BE, Bartlett PF (1999) Cellular distribution and developmental expression of AMP-activated protein kinase isoforms in mouse central nervous system. J Neurochem 72:1707–1716

    Article  PubMed  CAS  Google Scholar 

  89. Fryer LG, Parbu-Patel A, Carling D (2002) The Anti-diabetic drugs rosiglitazone and metformin stimulate AMP-activated protein kinase through distinct signaling pathways. J Biol Chem 277:25226–25232

    Article  PubMed  CAS  Google Scholar 

  90. Woods A, Dickerson K, Heath R, Hong SP, Momcilovic M, Johnstone SR, Carlson M, Carling D (2005) Ca2+/calmodulin-dependent protein kinase kinase-beta acts upstream of AMP-activated protein kinase in mammalian cells. Cell Metab 2:21–33

    Article  PubMed  CAS  Google Scholar 

  91. Hawley SA, Pan DA, Mustard KJ, Ross L, Bain J, Edelman AM, Frenguelli BG, Hardie DG (2005) Calmodulin-dependent protein kinase kinase-beta is an alternative upstream kinase for AMP-activated protein kinase. Cell Metab 2:9–19

    Article  PubMed  CAS  Google Scholar 

  92. Raney MA, Turcotte LP (2008) Evidence for the involvement of CaMKII and AMPK in Ca2+-dependent signaling pathways regulating FA uptake and oxidation in contracting rodent muscle. J Appl Physiol 104:1366–1373

    Article  PubMed  CAS  Google Scholar 

  93. Suzuki A, Kusakai G, Kishimoto A, Shimojo Y, Ogura T, Lavin MF, Esumi H (2004) IGF-1 phosphorylates AMPK-alpha subunit in ATM-dependent and LKB1-independent manner. Biochem Biophys Res Commun 324:986–992

    Article  PubMed  CAS  Google Scholar 

  94. Xie M, Zhang D, Dyck JR, Li Y, Zhang H, Morishima M, Mann DL, Taffet GE, Baldini A, Khoury DS, Schneider MD (2006) A pivotal role for endogenous TGF-beta-activated kinase-1 in the LKB1/AMP-activated protein kinase energy-sensor pathway. Proc Natl Acad Sci USA 103:17378–17383

    Article  PubMed  CAS  Google Scholar 

  95. Hurley RL, Barre LK, Wood SD, Anderson KA, Kemp BE, Means AR, Witters LA (2006) Regulation of AMP-activated protein kinase by multisite phosphorylation in response to agents that elevate cellular cAMP. J Biol Chem 281:36662–36672

    Article  PubMed  CAS  Google Scholar 

  96. Djouder N, Tuerk RD, Suter M, Salvioni P, Thali RF, Scholz R, Vaahtomeri K, Auchli Y, Rechsteiner H, Brunisholz RA, Viollet B, Makela TP, Wallimann T, Neumann D, Krek W (2010) PKA phosphorylates and inactivates AMPKalpha to promote efficient lipolysis. EMBO J 29:469–481

    Article  PubMed  CAS  Google Scholar 

  97. Lim CT, Kola B, Korbonits M (2010) AMPK as a mediator of hormonal signalling. J Mol Endocrinol 44:87–97

    Article  PubMed  CAS  Google Scholar 

  98. Hong YH, Varanasi US, Yang W, Leff T (2003) AMP-activated protein kinase regulates HNF4alpha transcriptional activity by inhibiting dimer formation and decreasing protein stability. J Biol Chem 278:27495–27501

    Article  PubMed  CAS  Google Scholar 

  99. Tsuboi T, da Silva Xavier G, Leclerc I, Rutter GA (2003) 5′-AMP-activated protein kinase controls insulin-containing secretory vesicle dynamics. J Biol Chem 278:52042–52051

    Google Scholar 

  100. Cai Y, Martens GA, Hinke SA, Heimberg H, Pipeleers D, Van de Casteele M (2007) Increased oxygen radical formation and mitochondrial dysfunction mediate beta cell apoptosis under conditions of AMP-activated protein kinase stimulation. Free Radic Biol Med 42:64–78

    Article  PubMed  CAS  Google Scholar 

  101. Kefas BA, Cai Y, Kerckhofs K, Ling Z, Martens G, Heimberg H, Pipeleers D, Van de Casteele M (2004) Metformin-induced stimulation of AMP-activated protein kinase in beta-cells impairs their glucose responsiveness and can lead to apoptosis. Biochem Pharmacol 68:409–416

    Article  PubMed  CAS  Google Scholar 

  102. Wang W, Yang X, Kawai T, Lopez de Silanes I, Mazan-Mamczarz K, Chen P, Chook YM, Quensel C, Kohler M, Gorospe M (2004) AMP-activated protein kinase-regulated phosphorylation and acetylation of importin alpha1: involvement in the nuclear import of RNA-binding protein HuR. J Biol Chem 279:48376-48388

    Google Scholar 

  103. Amato S, Liu X, Zheng B, Cantley L, Rakic P, Man HY (2011) AMP-activated protein kinase regulates neuronal polarization by interfering with PI 3-kinase localization. Science 332:247–251

    Article  PubMed  CAS  Google Scholar 

  104. Cai Y, Wang Q, Ling Z, Pipeleers D, McDermott P, Pende M, Heimberg H, Van de Casteele M (2008) Akt activation protects pancreatic beta cells from AMPK-mediated death through stimulation of mTOR. Biochem Pharmacol 75:1981–1993

    Article  PubMed  CAS  Google Scholar 

  105. Okoshi R, Ozaki T, Yamamoto H, Ando K, Koida N, Ono S, Koda T, Kamijo T, Nakagawara A, Kizaki H (2008) Activation of AMP-activated protein kinase induces p53-dependent apoptotic cell death in response to energetic stress. J Biol Chem 283:3979–3987

    Article  PubMed  CAS  Google Scholar 

  106. Li J, Jiang P, Robinson M, Lawrence TS, Sun Y (2003) AMPK-beta1 subunit is a p53-independent stress responsive protein that inhibits tumor cell growth upon forced expression. Carcinogenesis 24:827–834

    Article  PubMed  CAS  Google Scholar 

  107. Borger DR, Gavrilescu LC, Bucur MC, Ivan M, Decaprio JA (2008) AMP-activated protein kinase is essential for survival in chronic hypoxia. Biochem Biophys Res Commun 370:230–234

    Article  PubMed  CAS  Google Scholar 

  108. Culmsee C, Monnig J, Kemp BE, Mattson MP (2001) AMP-activated protein kinase is highly expressed in neurons in the developing rat brain and promotes neuronal survival following glucose deprivation. J Mol Neurosci 17:45–58

    Article  PubMed  CAS  Google Scholar 

  109. Terunuma M, Vargas KJ, Wilkins ME, Ramirez OA, Jaureguiberry-Bravo M, Pangalos MN, Smart TG, Moss SJ, Couve A (2010) Prolonged activation of NMDA receptors promotes dephosphorylation and alters postendocytic sorting of GABAB receptors. Proc Natl Acad Sci USA 107:13918–13923

    Article  PubMed  CAS  Google Scholar 

  110. Kuramoto N, Wilkins ME, Fairfax BP, Revilla-Sanchez R, Terunuma M, Tamaki K, Iemata M, Warren N, Couve A, Calver A, Horvath Z, Freeman K, Carling D, Huang L, Gonzales C, Cooper E, Smart TG, Pangalos MN, Moss SJ (2007) Phospho-dependent functional modulation of GABA(B) receptors by the metabolic sensor AMP-dependent protein kinase. Neuron 53:233–247

    Article  PubMed  CAS  Google Scholar 

  111. Choi JS, Park C, Jeong JW (2010) AMP-activated protein kinase is activated in Parkinson’s disease models mediated by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Biochem Biophys Res Commun 391:147–151

    Article  PubMed  CAS  Google Scholar 

  112. McCullough LD, Zeng Z, Li H, Landree LE, McFadden J, Ronnett GV (2005) Pharmacological inhibition of AMP-activated protein kinase provides neuroprotection in stroke. J Biol Chem 280:20493–20502

    Article  PubMed  CAS  Google Scholar 

  113. Lopez–Lopez C, Dietrich MO, Metzger F, Loetscher H, Torres-Aleman I (2007) Disturbed cross talk between insulin-like growth factor I and AMP-activated protein kinase as a possible cause of vascular dysfunction in the amyloid precursor protein/presenilin 2 mouse model of Alzheimer’s disease. J Neurosci 27:824–831

    Article  PubMed  CAS  Google Scholar 

  114. Shin E, Shin S, Kong H, Lee S, Do SG, Jo TH, Park YI, Lee CK, Hwang IK, Kim K (2011) Dietary aloe reduces adipogenesis via the activation of AMPK and suppresses obesity-related inflammation in obese mice. Immune Netw 11:107–113

    Article  PubMed  Google Scholar 

  115. Thornton C, Bright NJ, Sastre M, Muckett PJ, Carling D (2011) AMP-activated protein kinase (AMPK) is a tau kinase, activated in response to amyloid beta-peptide exposure. Biochem J 434:503–512

    Article  PubMed  CAS  Google Scholar 

  116. Vingtdeux V, Davies P, Dickson DW, Marambaud P (2011) AMPK is abnormally activated in tangle- and pre-tangle-bearing neurons in Alzheimer’s disease and other tauopathies. Acta Neuropathol 121:337–349

    Article  PubMed  CAS  Google Scholar 

  117. Salminen A, Kaarniranta K, Haapasalo A, Soininen H, Hiltunen M (2011) AMP-activated protein kinase: a potential player in Alzheimer’s disease. J Neurochem 118:460–474

    Article  PubMed  CAS  Google Scholar 

  118. Lim MA, Selak MA, Xiang Z, Krainc D, Neve RL, Kraemer BC, Watts JL, Kalb RG (2012) Reduced activity of AMP-activated protein kinase protects against genetic models of motor neuron disease. J Neurosci 32:1123–1141

    Article  PubMed  CAS  Google Scholar 

  119. Li J, Benashski S, McCullough LD (2011) Post-stroke hypothermia provides neuroprotection through inhibition of AMP-activated protein kinase. J Neurotrauma 28:1281–1288

    Article  PubMed  Google Scholar 

  120. Li J, Benashski SE, Siegel C, Liu F, McCullough LD (2010) Adenosine monophosphate activated protein kinase inhibition is protective in both sexes after experimental stroke. Neurosci Lett 482:62–65

    Article  PubMed  CAS  Google Scholar 

  121. Li J, Zeng Z, Viollet B, Ronnett GV, McCullough LD (2007) Neuroprotective effects of adenosine monophosphate-activated protein kinase inhibition and gene deletion in stroke. Stroke 38:2992–2999

    Article  PubMed  CAS  Google Scholar 

  122. Li J, Benashski SE, Venna VR, McCullough LD (2010) Effects of metformin in experimental stroke. Stroke 41:2645–2652

    Article  PubMed  CAS  Google Scholar 

  123. Son SM, Jung ES, Shin HJ, Byun J, Mook-Jung I (2012) Abeta-induced formation of autophagosomes is mediated by RAGE-CaMKKbeta-AMPK signaling. Neurobiol Aging 33(1006):e1011–e1023

    Google Scholar 

  124. Chen Y, Zhou K, Wang R, Liu Y, Kwak YD, Ma T, Thompson RC, Zhao Y, Smith L, Gasparini L, Luo Z, Xu H, Liao FF (2009) Antidiabetic drug metformin (GlucophageR) increases biogenesis of Alzheimer’s amyloid peptides via up-regulating BACE1 transcription. Proc Natl Acad Sci USA 106:3907–3912

    Article  PubMed  CAS  Google Scholar 

  125. Greco SJ, Sarkar S, Johnston JM, Tezapsidis N (2009) Leptin regulates tau phosphorylation and amyloid through AMPK in neuronal cells. Biochem Biophys Res Commun 380:98–104

    Article  PubMed  CAS  Google Scholar 

  126. Vingtdeux V, Giliberto L, Zhao H, Chandakkar P, Wu Q, Simon JE, Janle EM, Lobo J, Ferruzzi MG, Davies P, Marambaud P (2010) AMP-activated protein kinase signaling activation by resveratrol modulates amyloid-beta peptide metabolism. J Biol Chem 285:9100–9113

    Article  PubMed  CAS  Google Scholar 

  127. Vingtdeux V, Chandakkar P, Zhao H, d’Abramo C, Davies P, Marambaud P (2011) Novel synthetic small-molecule activators of AMPK as enhancers of autophagy and amyloid-beta peptide degradation. FASEB J 25:219–231

    Article  PubMed  CAS  Google Scholar 

  128. Won JS, Im YB, Kim J, Singh AK, Singh I (2010) Involvement of AMP-activated-protein-kinase (AMPK) in neuronal amyloidogenesis. Biochem Biophys Res Commun 399:487–491

    Article  PubMed  CAS  Google Scholar 

  129. Chou S-Y, Lee Y-C, Chen H-M, Chiang M-C, Lai H-L, Chang H–H, Wu Y-C, Sun C-N, Chien C-L, Lin Y-S, Wang S-C, Tung Y–Y, Chang C, Chern Y (2005) CGS21680 attenuates symptoms of Huntington’s disease in a transgenic mouse model. J Neurochem 93:310–320

    Article  PubMed  CAS  Google Scholar 

  130. She P, Zhang Z, Marchionini D, Diaz WC, Jetton TJ, Kimball SR, Vary TC, Lang CH, Lynch CJ (2011) Molecular characterization of skeletal muscle atrophy in the R6/2 mouse model of Huntington’s disease. Am J Physiol Endocrinol Metab 301:E49–E61

    Article  PubMed  CAS  Google Scholar 

  131. Chaturvedi RK, Calingasan NY, Yang L, Hennessey T, Johri A, Beal MF (2010) Impairment of PGC-1alpha expression, neuropathology and hepatic steatosis in a transgenic mouse model of Huntington’s disease following chronic energy deprivation. Hum Mol Genet 19:3190–3205

    Article  PubMed  CAS  Google Scholar 

  132. Ma TC, Buescher JL, Oatis B, Funk JA, Nash AJ, Carrier RL, Hoyt KR (2007) Metformin therapy in a transgenic mouse model of Huntington’s disease. Neurosci Lett 411:98–103

    Article  PubMed  CAS  Google Scholar 

  133. Wallimann T (1994) Bioenergetics. Dissecting the role of creatine kinase. Curr Biol 4:42–46

    Article  PubMed  CAS  Google Scholar 

  134. Wallimann T, Walzthony D, Wegmann G, Moser H, Eppenberger HM, Barrantes FJ (1985) Subcellular localization of creatine kinase in Torpedo electrocytes: association with acetylcholine receptor-rich membranes. J Cell Biol 100:1063–1072

    Article  PubMed  CAS  Google Scholar 

  135. Wyss M, Kaddurah-Daouk R (2000) Creatine and creatinine metabolism. Physiol Rev 80:1107–1213

    PubMed  CAS  Google Scholar 

  136. Wallimann T, Wyss M, Brdiczka D, Nicolay K, Eppenberger HM (1992) Intracellular compartmentation, structure and function of creatine kinase isoenzymes in tissues with high and fluctuating energy demands: the ‘phosphocreatine circuit’ for cellular energy homeostasis. Biochem J 281(Pt 1):21–40

    PubMed  CAS  Google Scholar 

  137. Linton JD, Holzhausen LC, Babai N, Song H, Miyagishima KJ, Stearns GW, Lindsay K, Wei J, Chertov AO, Peters TA, Caffe R, Pluk H, Seeliger MW, Tanimoto N, Fong K, Bolton L, Kuok DL, Sweet IR, Bartoletti TM, Radu RA, Travis GH, Zagotta WN, Townes-Anderson E, Parker E, Van der Zee CE, Sampath AP, Sokolov M, Thoreson WB, Hurley JB (2010) Flow of energy in the outer retina in darkness and in light. Proc Natl Acad Sci USA 107:8599–8604

    Article  PubMed  CAS  Google Scholar 

  138. Shin JB, Streijger F, Beynon A, Peters T, Gadzala L, McMillen D, Bystrom C, Van der Zee CE, Wallimann T, Gillespie PG (2007) Hair bundles are specialized for ATP delivery via creatine kinase. Neuron 53:371–386

    Article  PubMed  CAS  Google Scholar 

  139. Wallimann T, Hemmer W (1994) Creatine kinase in non-muscle tissues and cells. Mol Cell Biochem 133–134:193–220

    Article  PubMed  Google Scholar 

  140. Mahajan VB, Pai KS, Lau A, Cunningham DD (2000) Creatine kinase, an ATP-generating enzyme, is required for thrombin receptor signaling to the cytoskeleton. Proc Natl Acad Sci USA 97:12062–12067

    Article  PubMed  CAS  Google Scholar 

  141. Suidan HS, Nobes CD, Hall A, Monard D (1997) Astrocyte spreading in response to thrombin and lysophosphatidic acid is dependent on the Rho GTPase. Glia 21:244–252

    Article  PubMed  CAS  Google Scholar 

  142. Gurwitz D, Cunningham DD (1988) Thrombin modulates and reverses neuroblastoma neurite outgrowth. Proc Natl Acad Sci USA 85:3440–3444

    Article  PubMed  CAS  Google Scholar 

  143. Suidan HS, Stone SR, Hemmings BA, Monard D (1992) Thrombin causes neurite retraction in neuronal cells through activation of cell surface receptors. Neuron 8:363–375

    Article  PubMed  CAS  Google Scholar 

  144. Vaughan PJ, Pike CJ, Cotman CW, Cunningham DD (1995) Thrombin receptor activation protects neurons and astrocytes from cell death produced by environmental insults. J Neurosci 15:5389–5401

    PubMed  CAS  Google Scholar 

  145. Inoue K, Ueno S, Fukuda A (2004) Interaction of neuron-specific K+–Cl cotransporter, KCC2, with brain-type creatine kinase. FEBS Lett 564:131–135

    Article  PubMed  CAS  Google Scholar 

  146. Salin-Cantegrel A, Shekarabi M, Holbert S, Dion P, Rochefort D, Laganiere J, Dacal S, Hince P, Karemera L, Gaspar C, Lapointe JY, Rouleau GA (2008) HMSN/ACC truncation mutations disrupt brain-type creatine kinase-dependant activation of K +/Cl- cotransporter 3. Hum Mol Genet 17:2703–2711

    Article  PubMed  CAS  Google Scholar 

  147. Gulacsi A, Lee CR, Sik A, Viitanen T, Kaila K, Tepper JM, Freund TF (2003) Cell type-specific differences in chloride-regulatory mechanisms and GABA(A) receptor-mediated inhibition in rat substantia nigra. J Neurosci 23:8237–8246

    PubMed  CAS  Google Scholar 

  148. Race JE, Makhlouf FN, Logue PJ, Wilson FH, Dunham PB, Holtzman EJ (1999) Molecular cloning and functional characterization of KCC3, a new K–Cl cotransporter. Am J Physiol 277:C1210–C1219

    PubMed  CAS  Google Scholar 

  149. Howard HC, Mount DB, Rochefort D, Byun N, Dupre N, Lu J, Fan X, Song L, Riviere JB, Prevost C, Horst J, Simonati A, Lemcke B, Welch R, England R, Zhan FQ, Mercado A, Siesser WB, George AL Jr, McDonald MP, Bouchard JP, Mathieu J, Delpire E, Rouleau GA (2002) The K–Cl cotransporter KCC3 is mutant in a severe peripheral neuropathy associated with agenesis of the corpus callosum. Nat Genet 32:384–392

    Article  PubMed  CAS  Google Scholar 

  150. Dupre N, Howard HC, Mathieu J, Karpati G, Vanasse M, Bouchard JP, Carpenter S, Rouleau GA (2003) Hereditary motor and sensory neuropathy with agenesis of the corpus callosum. Ann Neurol 54:9–18

    Article  PubMed  Google Scholar 

  151. Lang F, Busch GL, Ritter M, Volkl H, Waldegger S, Gulbins E, Haussinger D (1998) Functional significance of cell volume regulatory mechanisms. Physiol Rev 78:247–306

    PubMed  CAS  Google Scholar 

  152. Rivera C, Voipio J, Payne JA, Ruusuvuori E, Lahtinen H, Lamsa K, Pirvola U, Saarma M, Kaila K (1999) The K+/Cl co-transporter KCC2 renders GABA hyperpolarizing during neuronal maturation. Nature 397:251–255

    Article  PubMed  CAS  Google Scholar 

  153. Aksenov MY, Aksenova MV, Payne RM, Smith CD, Markesbery WR, Carney JM (1997) The expression of creatine kinase isoenzymes in neocortex of patients with neurodegenerative disorders: Alzheimer’s and Pick’s disease. Exp Neurol 146:458–465

    Article  PubMed  CAS  Google Scholar 

  154. Aksenova MV, Aksenov MY, Payne RM, Trojanowski JQ, Schmidt ML, Carney JM, Butterfield DA, Markesbery WR (1999) Oxidation of cytosolic proteins and expression of creatine kinase BB in frontal lobe in different neurodegenerative disorders. Dement Geriatr Cogn Disord 10:158–165

    Article  PubMed  CAS  Google Scholar 

  155. Castegna A, Aksenov M, Aksenova M, Thongboonkerd V, Klein JB, Pierce WM, Booze R, Markesbery WR, Butterfield DA (2002) Proteomic identification of oxidatively modified proteins in Alzheimer’s disease brain. Part I: creatine kinase BB, glutamine synthase, and ubiquitin carboxy-terminal hydrolase L-1. Free Radic Biol Med 33:562–571

    Article  PubMed  CAS  Google Scholar 

  156. Aksenov M, Aksenova M, Butterfield DA, Markesbery WR (2000) Oxidative modification of creatine kinase BB in Alzheimer’s disease brain. J Neurochem 74:2520–2527

    Article  PubMed  CAS  Google Scholar 

  157. Sorolla MA, Reverter-Branchat G, Tamarit J, Ferrer I, Ros J, Cabiscol E (2008) Proteomic and oxidative stress analysis in human brain samples of Huntington disease. Free Radic Biol Med 45:667–678

    Article  PubMed  CAS  Google Scholar 

  158. Perluigi M, Poon HF, Maragos W, Pierce WM, Klein JB, Calabrese V, Cini C, De Marco C, Butterfield DA (2005) Proteomic analysis of protein expression and oxidative modification in r6/2 transgenic mice: a model of Huntington disease. Mol Cell Proteomics 4:1849–1861

    Article  PubMed  CAS  Google Scholar 

  159. Kim J, Amante DJ, Moody JP, Edgerly CK, Bordiuk OL, Smith K, Matson SA, Matson WR, Scherzer CR, Rosas HD, Hersch SM, Ferrante RJ (2010) Reduced creatine kinase as a central and peripheral biomarker in Huntington’s disease. Biochim Biophys Acta 1802:673–681

    Article  PubMed  CAS  Google Scholar 

  160. Deschepper M, Hoogendoorn B, Brooks S, Dunnett SB, Jones L (2011) Proteomic changes in the brains of Huntington’s disease mouse models reflect pathology and implicate mitochondrial changes. Brain Res Bull http://dx.doi.org/10.1016/j.brainresbull.2011.01.012

  161. Luthi-Carter R, Hanson SA, Strand AD, Bergstrom DA, Chun W, Peters NL, Woods AM, Chan EY, Kooperberg C, Krainc D, Young AB, Tapscott SJ, Olson JM (2002) Dysregulation of gene expression in the R6/2 model of polyglutamine disease: parallel changes in muscle and brain. Hum Mol Genet 11:1911–1926

    Article  PubMed  CAS  Google Scholar 

  162. Zhang SF, Hennessey T, Yang L, Starkova NN, Beal MF, Starkov AA (2011) Impaired brain creatine kinase activity in Huntington’s disease. Neurodegener Dis 8:194–201

    Article  PubMed  CAS  Google Scholar 

  163. Mochel F, Durant B, Meng X, O’Callaghan J, Yu H, Brouillet E, Wheeler VC, Humbert S, Schiffmann R, Durr A (2012) Early alterations of brain cellular energy homeostasis in Huntington disease models. J Biol Chem 287:1361–1370

    Article  PubMed  CAS  Google Scholar 

  164. Li H, Khirug S, Cai C, Ludwig A, Blaesse P, Kolikova J, Afzalov R, Coleman SK, Lauri S, Airaksinen MS, Keinanen K, Khiroug L, Saarma M, Kaila K, Rivera C (2007) KCC2 interacts with the dendritic cytoskeleton to promote spine development. Neuron 56:1019–1033

    Article  PubMed  CAS  Google Scholar 

  165. Spires TL, Grote HE, Garry S, Cordery PM, Van Dellen A, Blakemore C, Hannan AJ (2004) Dendritic spine pathology and deficits in experience-dependent dendritic plasticity in R6/1 Huntington’s disease transgenic mice. Eur J Neurosci 19:2799–2807

    Article  PubMed  Google Scholar 

  166. Choi H, Park CS, Kim BG, Cho JW, Park JB, Bae YS, Bae DS (2001) Creatine kinase B is a target molecule of reactive oxygen species in cervical cancer. Mol Cells 12:412–417

    PubMed  CAS  Google Scholar 

  167. Mekhfi H, Veksler V, Mateo P, Maupoil V, Rochette L, Ventura-Clapier R (1996) Creatine kinase is the main target of reactive oxygen species in cardiac myofibrils. Circ Res 78:1016–1027

    Article  PubMed  CAS  Google Scholar 

  168. Firdaus WJ, Wyttenbach A, Giuliano P, Kretz-Remy C, Currie RW, Arrigo AP (2006) Huntingtin inclusion bodies are iron-dependent centers of oxidative events. FEBS J 273:5428–5441

    Article  PubMed  CAS  Google Scholar 

  169. Lawler JM, Barnes WS, Wu G, Song W, Demaree S (2002) Direct antioxidant properties of creatine. Biochem Biophys Res Commun 290:47–52

    Article  PubMed  CAS  Google Scholar 

  170. Klopstock T, Elstner M, Bender A (2011) Creatine in mouse models of neurodegeneration and aging. Amino Acids 40:1297–1303

    Article  PubMed  CAS  Google Scholar 

  171. Ferrante RJ, Andreassen OA, Jenkins BG, Dedeoglu A, Kuemmerle S, Kubilus JK, Kaddurah-Daouk R, Hersch SM, Beal MF (2000) Neuroprotective effects of creatine in a transgenic mouse model of Huntington’s disease. J Neurosci 20:4389–4397

    PubMed  CAS  Google Scholar 

  172. Matthews RT, Ferrante RJ, Klivenyi P, Yang L, Klein AM, Mueller G, Kaddurah-Daouk R, Beal MF (1999) Creatine and cyclocreatine attenuate MPTP neurotoxicity. Exp Neurol 157:142–149

    Article  PubMed  CAS  Google Scholar 

  173. Klivenyi P, Ferrante RJ, Matthews RT, Bogdanov MB, Klein AM, Andreassen OA, Mueller G, Wermer M, Kaddurah-Daouk R, Beal MF (1999) Neuroprotective effects of creatine in a transgenic animal model of amyotrophic lateral sclerosis. Nat Med 5:347–350

    Article  PubMed  CAS  Google Scholar 

  174. Andres RH, Ducray AD, Schlattner U, Wallimann T, Widmer HR (2008) Functions and effects of creatine in the central nervous system. Brain Res Bull 76:329–343

    Article  PubMed  CAS  Google Scholar 

  175. Perasso L, Cupello A, Lunardi GL, Principato C, Gandolfo C, Balestrino M (2003) Kinetics of creatine in blood and brain after intraperitoneal injection in the rat. Brain Res 974:37–42

    Article  PubMed  CAS  Google Scholar 

  176. Bae BI, Xu H, Igarashi S, Fujimuro M, Agrawal N, Taya Y, Hayward SD, Moran TH, Montell C, Ross CA, Snyder SH, Sawa A (2005) p53 mediates cellular dysfunction and behavioral abnormalities in Huntington’s disease. Neuron 47:29–41

    Article  PubMed  CAS  Google Scholar 

  177. Zhao J, Schmieg FI, Simmons DT, Molloy GR (1994) Mouse p53 represses the rat brain creatine kinase gene but activates the rat muscle creatine kinase gene. Mol Cell Biol 14:8483–8492

    PubMed  CAS  Google Scholar 

  178. Zhao TJ, Yan YB, Liu Y, Zhou HM (2007) The generation of the oxidized form of creatine kinase is a negative regulation on muscle creatine kinase. J Biol Chem 282:12022–12029

    Article  PubMed  CAS  Google Scholar 

  179. Matthews RT, Yang L, Jenkins BG, Ferrante RJ, Rosen BR, Kaddurah-Daouk R, Beal MF (1998) Neuroprotective effects of creatine and cyclocreatine in animal models of Huntington’s disease. J Neurosci 18:156–163

    PubMed  CAS  Google Scholar 

  180. Inoue K, Yamada J, Ueno S, Fukuda A (2006) Brain-type creatine kinase activates neuron-specific K+–Cl co-transporter KCC2. J Neurochem 96:598–608

    Article  PubMed  CAS  Google Scholar 

  181. Stockler S, Isbrandt D, Hanefeld F, Schmidt B, von Figura K (1996) Guanidinoacetate methyltransferase deficiency: the first inborn error of creatine metabolism in man. Am J Hum Genet 58:914–922

    PubMed  CAS  Google Scholar 

  182. Item CB, Stockler-Ipsiroglu S, Stromberger C, Muhl A, Alessandri MG, Bianchi MC, Tosetti M, Fornai F, Cioni G (2001) Arginine:glycine amidinotransferase deficiency: the third inborn error of creatine metabolism in humans. Am J Hum Genet 69:1127–1133

    Article  PubMed  CAS  Google Scholar 

  183. Battini R, Leuzzi V, Carducci C, Tosetti M, Bianchi MC, Item CB, Stockler-Ipsiroglu S, Cioni G (2002) Creatine depletion in a new case with AGAT deficiency: clinical and genetic study in a large pedigree. Mol Genet Metab 77:326–331

    Article  PubMed  CAS  Google Scholar 

  184. Salomons GS, van Dooren SJ, Verhoeven NM, Cecil KM, Ball WS, Degrauw TJ, Jakobs C (2001) X-linked creatine-transporter gene (SLC6A8) defect: a new creatine-deficiency syndrome. Am J Hum Genet 68:1497–1500

    Article  PubMed  CAS  Google Scholar 

  185. Bizzi A, Bugiani M, Salomons GS, Hunneman DH, Moroni I, Estienne M, Danesi U, Jakobs C, Uziel G (2002) X-linked creatine deficiency syndrome: a novel mutation in creatine transporter gene SLC6A8. Ann Neurol 52:227–231

    Article  PubMed  CAS  Google Scholar 

  186. Brookmeyer R, Gray S, Kawas C (1998) Projections of Alzheimer’s disease in the United States and the public health impact of delaying disease onset. Am J Public Health 88:1337–1342

    Article  PubMed  CAS  Google Scholar 

  187. Zesiewicz TA, Baker MJ, Dunne PB, Hauser RA (2001) Diffuse Lewy Body Disease. Curr Treat Options Neurol 3:507–518

    Article  PubMed  Google Scholar 

  188. Gaig C, Valldeoriola F, Gelpi E, Ezquerra M, Llufriu S, Buongiorno M, Rey MJ, Marti MJ, Graus F, Tolosa E (2011) Rapidly progressive diffuse Lewy body disease. Mov Disord 26:1316–1323

    Article  PubMed  Google Scholar 

  189. Walker FO (2007) Huntington’s disease. Semin Neurol 27:143–150

    Article  PubMed  Google Scholar 

  190. Zhang SF, Hennessey T, Yang L, Starkova NN, Beal MF, Starkov AA (2010) Impaired brain creatine kinase activity in Huntington’s disease. Neurodegener Dis 8:194–201

    Article  PubMed  CAS  Google Scholar 

  191. Boettger T, Rust MB, Maier H, Seidenbecher T, Schweizer M, Keating DJ, Faulhaber J, Ehmke H, Pfeffer C, Scheel O, Lemcke B, Horst J, Leuwer R, Pape HC, Volkl H, Hubner CA, Jentsch TJ (2003) Loss of K–Cl co-transporter KCC3 causes deafness, neurodegeneration and reduced seizure threshold. EMBO J 22:5422–5434

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

We thank Mr. D.P. Chamberlin for reading and editing the manuscript. This work was supported by grants from Academia Sinica (AS-97-TP-B02, AS-100-TP2-B02), the Institute of Biomedical Sciences/Academia Sinica (Clinical Research Center grants, CRC98-P03B; CRC101-P02), and the National Science Council (NSC97-2321-B-001-030; NSC 99-2321-B-001-012), Taipei, Taiwan.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yijuang Chern.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ju, TC., Lin, YS. & Chern, Y. Energy dysfunction in Huntington’s disease: insights from PGC-1α, AMPK, and CKB. Cell. Mol. Life Sci. 69, 4107–4120 (2012). https://doi.org/10.1007/s00018-012-1025-2

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-012-1025-2

Keywords

Navigation