Skip to main content

Mitochondrial Dysfunction in Huntington’s Disease

  • Chapter
  • First Online:
Polyglutamine Disorders

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 1049))

Abstract

Mitochondrial dysfunction has been described as an early pathological mechanism delineating the selective neurodegeneration that occurs in Huntington’s disease (HD), a polyglutamine-expansion disorder that largely affects the striatum and the cerebral cortex. Over the years, mitochondria roles in eukaryotic cells (e.g. in neurons) have largely diverged from the classically attributed cell power source; indeed, mitochondria not only contribute for synthesis of several metabolites, but are also dynamic organelles that fragment and fuse to achieve a maximal bioenergetic performance, are transported along microtubules, regulate intracellular calcium homeostasis through the interaction with the endoplasmic reticulum, produce free radicals and participate in cell death processes. Indeed, most of these activities have been demonstrated to be affected in HD, potentially contributing for the neuronal dysfunction in pre-symptomatic stages. This chapter resumes some of the evidences that pose mitochondria as a main regulatory organelle in HD-affected neurons, uncovering some potentially therapeutic mitochondrial-based relevant targets.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 109.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 139.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 199.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Abbreviations

Δψm:

Mitochondrial membrane potential

α-KGDH:

α-ketoglutarate dehydrogenase

3-NP:

3-nitropropionic acid

AIF:

Apoptosis inducing factor

Apaf-1:

Apoptotic protease-activating factor 1

ATP:

Adenosine triphosphate

Bcl-2:

B-cell lymphoma 2

BDNF:

Brain derived neurotrophic factor

BH3:

Bcl-2 homology 3

Bid:

BH3 interacting-domain death agonist

Bim:

Bcl-2 interacting mediator of cell death

BNIP3:

BCL2/adenovirus E1B 19 kDa protein-interacting protein 3

CBP:

CREB-binding protein

CK:

Creatine kinase

CoQ:

Coenzyme Q

CREB:

cAMP response element-binding protein

Drp1:

Dynamin-related protein 1

ETC:

Electron transport chain

Fis1:

Mitochondrial fission 1

FMN:

Flavin mononucleotide

GABA:

γ-aminobutyric acid

GAPDH:

Glyceraldehyde-3-phosphate dehydrogenase

Gpx:

Glutathione peroxidases

GTP:

Guanosine triphosphate

H2O2:

Hydrogen peroxide

HD:

Huntington’s disease

hESC:

Human embryonic stem cells

HTT/HTT:

Human huntingtin protein/gene

Htt:

Rodent huntingtin protein

IAP1:

Inhibitor of Apoptosis Protein-1

iPSCs:

Induced pluripotent stem cells

K:

Lysine

LC3:

Light chain 3

MCU:

Mitochondrial calcium uniporter

Mff:

Mitochondrial fission factor

Mfn:

Mitofusin

mHTT:

Human mutant HTT

mHtt:

Rodent mutant Htt

MIM:

Mitochondrial inner membrane

MIS:

Mitochondrial intermembrane space

MOM:

Mitochondrial outer membrane

mtDNA:

Mitochondrial DNA

NAD:

β-nicotinamide adenine dinucleotide

ND5:

NADH dehydrogenase subunit 5

NRF:

Nuclear respiratory factor

Nrf2:

Nuclear factor-erythroid 2-related factor-2

OPA1:

Optic atrophy 1

OXPHOS:

Oxidative phosphorylation

PCr:

Phosphocreatine

PDH:

Pyruvate dehydrogenase

PGC-1α:

PPARγ—coactivator-1α

PINK1:

PTEN-induced putative kinase 1

PolyQ:

Polyglutamine

PPAR:

Peroxisome proliferator-activated receptor

Prx:

Peroxiredoxins

PTEN:

Phosphatase and tensin homolog

PTP:

Permeability transition pore

PUMA:

p 53 upregulated modulator of apoptosis

ROS:

Reactive oxygen species

SDH:

Succinate dehydrogenase

Smac/DIABLO:

Second mitochondria derived activator of caspase/direct inhibitor of apoptosis-binding protein with low pI

SOD:

Superoxide dismutase

TAF:

TBP-associated factor 4

TBP:

TATA-binding protein

TCA:

Tricarboxylic acid

Tfam:

Mitochondrial transcription factor A

TIM:

Translocase of the inner membrane

TRAK:

Trafficking kinesin protein

XIAP:

X-linked inhibitor of apoptosis

YAC:

Yeast artificial chromosome

References

  1. Chaturvedi RK, Flint Beal M (2013) Mitochondrial diseases of the brain. Free Radic Biol Med 63:1–29

    Article  CAS  PubMed  Google Scholar 

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

    Google Scholar 

  3. Kim J, Moody JP, Edgerly CK, Bordiuk OL, Cormier K, Smith K, Beal MF, Ferrante RJ (2010) Mitochondrial loss, dysfunction and altered dynamics in Huntington’s disease. Hum Mol Genet 19:3919–3935

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kodsi MH, Swerdlow NR (1997) Mitochondrial toxin 3-nitropropionic acid produces startle reflex abnormalities and striatal damage in rats that model some features of Huntington’s disease. Neurosci Lett 231:103–107

    Article  CAS  PubMed  Google Scholar 

  5. Saft C, Zange J, Andrich J, Muller K, Lindenberg K, Landwehrmeyer B, Vorgerd M, Kraus PH, Przuntek H, Schols L (2005) Mitochondrial impairment in patients and asymptomatic mutation carriers of Huntington’s disease. Mov Disord Off J Mov Disord Soc 20:674–679

    Article  Google Scholar 

  6. Goebel HH, Heipertz R, Scholz W, Iqbal K, Tellez-Nagel I (1978) Juvenile Huntington chorea: clinical, ultrastructural, and biochemical studies. Neurology 28:23–31

    Article  CAS  PubMed  Google Scholar 

  7. Beal MF, Brouillet E, Jenkins BG, Ferrante RJ, Kowall NW, Miller JM, Storey E, Srivastava R, Rosen BR, Hyman BT (1993) Neurochemical and histologic characterization of striatal excitotoxic lesions produced by the mitochondrial toxin 3-nitropropionic acid. J Neurosci Off J Soc Neurosci 13:4181–4192

    Article  CAS  Google Scholar 

  8. Brouillet E, Hantraye P, Ferrante RJ, Dolan R, Leroy-Willig A, Kowall NW, Beal MF (1995) Chronic mitochondrial energy impairment produces selective striatal degeneration and abnormal choreiform movements in primates. Proc Natl Acad Sci USA 92:7105–7109

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. 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  CAS  PubMed  Google Scholar 

  10. Choo YS, Johnson GV, MacDonald M, Detloff PJ, Lesort M (2004) Mutant huntingtin directly increases susceptibility of mitochondria to the calcium-induced permeability transition and cytochrome c release. Hum Mol Genet 13:1407–1420

    Article  CAS  PubMed  Google Scholar 

  11. 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

    Article  CAS  PubMed  Google Scholar 

  12. Song W, Chen J, Petrilli A, Liot G, Klinglmayr E, Zhou Y, Poquiz P, Tjong J, Pouladi MA, Hayden MR et al (2011) Mutant huntingtin binds the mitochondrial fission GTPase dynamin-related protein-1 and increases its enzymatic activity. Nat Med 17:377–382

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Napoli E, Wong S, Hung C, Ross-Inta C, Bomdica P, Giulivi C (2013) Defective mitochondrial disulfide relay system, altered mitochondrial morphology and function in Huntington’s disease. Hum Mol Genet 22:989–1004

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  15. Yano H, Baranov SV, Baranova OV, Kim J, Pan Y, Yablonska S, Carlisle DL, Ferrante RJ, Kim AH, Friedlander RM (2014) Inhibition of mitochondrial protein import by mutant huntingtin. Nat Neurosci 17:822–831

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Nicholls DG, Ward MW (2000) Mitochondrial membrane potential and neuronal glutamate excitotoxicity: mortality and millivolts. Trends Neurosci 23(4):166–174

    Article  CAS  PubMed  Google Scholar 

  17. 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  CAS  PubMed  Google Scholar 

  18. Naia L, Ferreira IL, Cunha-Oliveira T, Duarte AI, Ribeiro M, Rosenstock TR, Laco MN, Ribeiro MJ, Oliveira CR, Saudou F et al (2015) Activation of IGF-1 and insulin signaling pathways ameliorate mitochondrial function and energy metabolism in Huntington’s Disease human lymphoblasts. Mol Neurobiol 51:331–348

    Article  CAS  PubMed  Google Scholar 

  19. Sawa A, Wiegand GW, Cooper J, Margolis RL, Sharp AH, Lawler JF Jr, Greenamyre JT, Snyder SH, Ross CA (1999) Increased apoptosis of Huntington disease lymphoblasts associated with repeat length-dependent mitochondrial depolarization. Nat Med 5:1194–1198

    Article  CAS  PubMed  Google Scholar 

  20. 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  CAS  PubMed  Google Scholar 

  21. Ferreira IL, Nascimento MV, Ribeiro M, Almeida S, Cardoso SM, Grazina M, Pratas J, Santos MJ, Januario C, Oliveira CR et al (2010) Mitochondrial-dependent apoptosis in Huntington’s disease human cybrids. Exp Neurol 222:243–255

    Article  CAS  PubMed  Google Scholar 

  22. Humbert S, Bryson EA, Cordelieres FP, Connors NC, Datta SR, Finkbeiner S, Greenberg ME, Saudou F (2002) The IGF-1/Akt pathway is neuroprotective in Huntington’s disease and involves Huntingtin phosphorylation by Akt. Dev Cell 2:831–837

    Article  CAS  PubMed  Google Scholar 

  23. Benchoua A, Trioulier Y, Zala D, Gaillard MC, Lefort N, Dufour N, Saudou F, Elalouf JM, Hirsch E, Hantraye P et al (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  CAS  PubMed  PubMed Central  Google Scholar 

  24. Browne SE (2008) Mitochondria and Huntington’s disease pathogenesis: insight from genetic and chemical models. Ann N Y Acad Sci 1147:358–382

    Article  CAS  PubMed  Google Scholar 

  25. Maksimovic ID, Jovanovic MD, Colic M, Mihajlovic R, Micic D, Selakovic V, Ninkovic M, Malicevic Z, Rusic-Stojiljkovic M, Jovicic A (2001) Oxidative damage and metabolic dysfunction in experimental Huntington’s disease: selective vulnerability of the striatum and hippocampus. Vojnosanit Pregl 58:237–242

    CAS  PubMed  Google Scholar 

  26. Pandey M, Varghese M, Sindhu KM, Sreetama S, Navneet AK, Mohanakumar KP, Usha R (2008) Mitochondrial NAD + -linked State 3 respiration and complex-I activity are compromised in the cerebral cortex of 3-nitropropionic acid-induced rat model of Huntington’s disease. J Neurochem 104:420–434

    CAS  PubMed  Google Scholar 

  27. Silva AC, Almeida S, Laco M, Duarte AI, Domingues J, Oliveira CR, Januario C, Rego AC (2013) Mitochondrial respiratory chain complex activity and bioenergetic alterations in human platelets derived from pre-symptomatic and symptomatic Huntington’s disease carriers. Mitochondrion 13:801–809

    Article  CAS  PubMed  Google Scholar 

  28. Chakraborty J, Rajamma U, Mohanakumar KP (2014) A mitochondrial basis for Huntington’s disease: therapeutic prospects. Mol Cell Biochem 389:277–291

    Article  CAS  PubMed  Google Scholar 

  29. Oliveira JM, Jekabsons MB, Chen S, Lin A, Rego AC, Goncalves J, Ellerby LM, Nicholls DG (2007) Mitochondrial dysfunction in Huntington’s disease: the bioenergetics of isolated and in situ mitochondria from transgenic mice. J Neurochem 101:241–249

    Article  CAS  PubMed  Google Scholar 

  30. Brustovetsky N, LaFrance R, Purl KJ, Brustovetsky T, Keene CD, Low WC, Dubinsky JM (2005) Age-dependent changes in the calcium sensitivity of striatal mitochondria in mouse models of Huntington’s Disease. J Neurochem 93:1361–1370

    Article  CAS  PubMed  Google Scholar 

  31. Pellman JJ, Hamilton J, Brustovetsky T, Brustovetsky N (2015) Ca(2+) handling in isolated brain mitochondria and cultured neurons derived from the YAC128 mouse model of Huntington’s disease. J Neurochem 134:652–667

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. HD iPSC Consortium (2012) Induced pluripotent stem cells from patients with Huntington’s disease show CAG-repeat-expansion-associated phenotypes. Cell stem cell 11:264–278

    Article  CAS  Google Scholar 

  33. Mochel F, N’Guyen TM, Deelchand D, Rinaldi D, Valabregue R, Wary C, Carlier PG, Durr A, Henry PG (2012) Abnormal response to cortical activation in early stages of Huntington disease. Movement disorders: official journal of the Movement Disorder Society 27:907–910

    Article  CAS  Google Scholar 

  34. Lopes C, Ribeiro M, Duarte AI, Humbert S, Saudou F, Pereira de Almeida L, Hayden M, Rego AC (2014) IGF-1 intranasal administration rescues Huntington’s disease phenotypes in YAC128 mice. Mol Neurobiol 49:1126–1142

    Article  CAS  PubMed  Google Scholar 

  35. 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  CAS  PubMed  Google Scholar 

  36. Seong IS, Ivanova E, Lee JM, Choo YS, Fossale E, Anderson M, Gusella JF, Laramie JM, Myers RH, Lesort M et al (2005) HD CAG repeat implicates a dominant property of huntingtin in mitochondrial energy metabolism. Hum Mol Genet 14:2871–2880

    Article  CAS  PubMed  Google Scholar 

  37. 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  CAS  PubMed  Google Scholar 

  38. Lodi R, Schapira AH, Manners D, Styles P, Wood NW, Taylor DJ, Warner TT (2000) Abnormal in vivo skeletal muscle energy metabolism in Huntington’s disease and dentatorubropallidoluysian atrophy. Ann Neurol 48:72–76

    Article  CAS  PubMed  Google Scholar 

  39. Naia L, Ribeiro M, Rodrigues J, Duarte AI, Lopes C, Rosenstock TR, Hayden MR, Rego AC (2016) Insulin and IGF-1 regularize energy metabolites in neural cells expressing full-length mutant huntingtin. Neuropeptides

    Google Scholar 

  40. Antonini A, Leenders KL, Spiegel R, Meier D, Vontobel P, Weigell-Weber M, Sanchez-Pernaute R, de Yebenez JG, Boesiger P, Weindl A et al (1996) Striatal glucose metabolism and dopamine D2 receptor binding in asymptomatic gene carriers and patients with Huntington’s disease. Brain A J Neurol 119(Pt 6):2085–2095

    Article  Google Scholar 

  41. Mazziotta JC, Phelps ME, Pahl JJ, Huang SC, Baxter LR, Riege WH, Hoffman JM, Kuhl DE, Lanto AB, Wapenski JA et al (1987) Reduced cerebral glucose metabolism in asymptomatic subjects at risk for Huntington’s disease. The New England journal of medicine 316:357–362

    Article  CAS  PubMed  Google Scholar 

  42. Powers WJ, Haas RH, Le T, Videen TO, Hershey T, McGee-Minnich L, Perlmutter JS (2007) Normal platelet mitochondrial complex I activity in Huntington’s disease. Neurobiology of disease 27:99–101

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Burke JR, Enghild JJ, Martin ME, Jou YS, Myers RM, Roses AD, Vance JM, Strittmatter WJ (1996) Huntingtin and DRPLA proteins selectively interact with the enzyme GAPDH. Nat Med 2:347–350

    Article  CAS  PubMed  Google Scholar 

  44. Jacobsen JC, Gregory GC, Woda JM, Thompson MN, Coser KR, Murthy V, Kohane IS, Gusella JF, Seong IS, MacDonald ME et al (2011) HD CAG-correlated gene expression changes support a simple dominant gain of function. Hum Mol Genet 20:2846–2860

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Jenkins BG, Koroshetz WJ, Beal MF, Rosen BR (1993) Evidence for impairment of energy metabolism in vivo in Huntington’s disease using localized 1H NMR spectroscopy. Neurology 43:2689–2695

    Article  CAS  PubMed  Google Scholar 

  46. Koroshetz WJ, Jenkins BG, Rosen BR, Beal MF (1997) Energy metabolism defects in Huntington’s disease and effects of coenzyme Q10. Ann Neurol 41:160–165

    Article  CAS  PubMed  Google Scholar 

  47. Martin WR, Wieler M, Hanstock CC (2007) Is brain lactate increased in Huntington’s disease? J Neurol Sci 263:70–74

    Article  CAS  PubMed  Google Scholar 

  48. Tsang TM, Woodman B, McLoughlin GA, Griffin JL, Tabrizi SJ, Bates GP, Holmes E (2006) Metabolic characterization of the R6/2 transgenic mouse model of Huntington’s disease by high-resolution MAS 1H NMR spectroscopy. J Proteome Res 5:483–492

    Article  CAS  PubMed  Google Scholar 

  49. Ferreira IL, Cunha-Oliveira T, Nascimento MV, Ribeiro M, Proenca MT, Januario C, Oliveira CR, Rego AC (2011) Bioenergetic dysfunction in Huntington’s disease human cybrids. Exp Neurol 231:127–134

    Article  CAS  PubMed  Google Scholar 

  50. Butterworth J, Yates CM, Reynolds GP (1985) Distribution of phosphate-activated glutaminase, succinic dehydrogenase, pyruvate dehydrogenase and gamma-glutamyl transpeptidase in post-mortem brain from Huntington’s disease and agonal cases. J Neurol Sci 67:161–171

    Article  CAS  PubMed  Google Scholar 

  51. Sorbi S, Bird ED, Blass JP (1983) Decreased pyruvate dehydrogenase complex activity in Huntington and Alzheimer brain. Ann Neurol 13:72–78

    Article  CAS  PubMed  Google Scholar 

  52. Andreassen OA, Ferrante RJ, Huang HM, Dedeoglu A, Park L, Ferrante KL, Kwon J, Borchelt DR, Ross CA, Gibson GE et al (2001) Dichloroacetate exerts therapeutic effects in transgenic mouse models of Huntington’s disease. Ann Neurol 50:112–117

    Article  CAS  PubMed  Google Scholar 

  53. Klivenyi P, Starkov AA, Calingasan NY, Gardian G, Browne SE, Yang L, Bubber P, Gibson GE, Patel MS, Beal MF (2004) Mice deficient in dihydrolipoamide dehydrogenase show increased vulnerability to MPTP, malonate and 3-nitropropionic acid neurotoxicity. J Neurochem 88:1352–1360

    Article  CAS  PubMed  Google Scholar 

  54. Tabrizi SJ, Cleeter MW, Xuereb J, Taanman JW, Cooper JM, Schapira AH (1999) Biochemical abnormalities and excitotoxicity in Huntington’s disease brain. Ann Neurol 45:25–32

    Article  CAS  PubMed  Google Scholar 

  55. Naseri NN, Xu H, Bonica J, Vonsattel JP, Cortes EP, Park LC, Arjomand J, Gibson GE (2015) Abnormalities in the tricarboxylic Acid cycle in Huntington disease and in a Huntington disease mouse model. J Neuropathol Exp Neurol 74:527–537

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Sorolla MA, Rodriguez-Colman MJ, Tamarit J, Ortega Z, Lucas JJ, Ferrer I, Ros J, Cabiscol E (2010) Protein oxidation in Huntington disease affects energy production and vitamin B6 metabolism. Free Radic Biol Med 49:612–621

    Article  CAS  PubMed  Google Scholar 

  57. Vinogradov AD, Grivennikova VG (2015) Oxidation of NADH and ROS production by respiratory complex I. Biochim Biophys Acta

    Google Scholar 

  58. Bleier L, Drose S (2013) Superoxide generation by complex III: from mechanistic rationales to functional consequences. Biochim Biophys Acta 1827:1320–1331

    Article  CAS  PubMed  Google Scholar 

  59. Murphy MP (2009) How mitochondria produce reactive oxygen species. Biochem J 417:1–13

    Article  CAS  PubMed  Google Scholar 

  60. Flynn JM, Melov S (2013) SOD2 in mitochondrial dysfunction and neurodegeneration. Free Radic Biol Med 62:4–12

    Article  CAS  PubMed  Google Scholar 

  61. Rabilloud T, Heller M, Rigobello MP, Bindoli A, Aebersold R, Lunardi J (2001) The mitochondrial antioxidant defence system and its response to oxidative stress. Proteomics 1:1105–1110

    Article  CAS  PubMed  Google Scholar 

  62. Hands S, Sajjad MU, Newton MJ, Wyttenbach A (2011) In vitro and in vivo aggregation of a fragment of huntingtin protein directly causes free radical production. J Biol Chem 286:44512–44520

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. 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  CAS  PubMed  Google Scholar 

  64. Tabrizi SJ, Schapira AH (1999) Secondary abnormalities of mitochondrial DNA associated with neurodegeneration. Biochem Soc Symp 66:99–110

    Article  CAS  PubMed  Google Scholar 

  65. Tabrizi SJ, Workman J, Hart PE, Mangiarini L, Mahal A, Bates G, Cooper JM, Schapira AH (2000) Mitochondrial dysfunction and free radical damage in the Huntington R6/2 transgenic mouse. Ann Neurol 47:80–86

    Article  CAS  PubMed  Google Scholar 

  66. Ribeiro M, Rosenstock TR, Oliveira AM, Oliveira CR, Rego AC (2014) Insulin and IGF-1 improve mitochondrial function in a PI-3 K/Akt-dependent manner and reduce mitochondrial generation of reactive oxygen species in Huntington’s disease knock-in striatal cells. Free Radic Biol Med 74:129–144

    Article  CAS  PubMed  Google Scholar 

  67. Ribeiro M, Rosenstock TR, Cunha-Oliveira T, Ferreira IL, Oliveira CR, Rego AC (2012) Glutathione redox cycle dysregulation in Huntington’s disease knock-in striatal cells. Free Radic Biol Med 53:1857–1867

    Article  CAS  PubMed  Google Scholar 

  68. Chae JI, Kim DW, Lee N, Jeon YJ, Jeon I, Kwon J, Kim J, Soh Y, Lee DS, Seo KS et al (2012) Quantitative proteomic analysis of induced pluripotent stem cells derived from a human Huntington’s disease patient. Biochem J 446:359–371

    Article  CAS  PubMed  Google Scholar 

  69. Stotland A, Gottlieb RA (2015) Mitochondrial quality control: easy come, easy go. Biochim Biophys Acta 1853:2802–2811

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Zhu J, Wang KZ, Chu CT (2013) After the banquet: mitochondrial biogenesis, mitophagy, and cell survival. Autophagy 9:1663–1676

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Jin YN, Johnson GV (2010) The interrelationship between mitochondrial dysfunction and transcriptional dysregulation in Huntington disease. J Bioenerg Biomembr 42:199–205

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Steffan JS, Kazantsev A, Spasic-Boskovic O, Greenwald M, Zhu YZ, Gohler H, Wanker EE, Bates GP, Housman DE, Thompson LM (2000) The Huntington’s disease protein interacts with p53 and CREB-binding protein and represses transcription. Proc Natl Acad Sci USA 97:6763–6768

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. 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  CAS  PubMed  Google Scholar 

  74. Johri A, Chandra A, Beal MF (2013) PGC-1alpha, mitochondrial dysfunction, and Huntington’s disease. Free Radic Biol Med 62:37–46

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Palikaras K, Tavernarakis N (2014) Mitochondrial homeostasis: the interplay between mitophagy and mitochondrial biogenesis. Exp Gerontol 56:182–188

    Article  CAS  PubMed  Google Scholar 

  76. Shirendeb UP, Calkins MJ, Manczak M, Anekonda V, Dufour B, McBride JL, Mao P, Reddy PH (2012) Mutant huntingtin’s interaction with mitochondrial protein Drp1 impairs mitochondrial biogenesis and causes defective axonal transport and synaptic degeneration in Huntington’s disease. Hum Mol Genet 21:406–420

    Article  CAS  PubMed  Google Scholar 

  77. Weydt P, Pineda VV, Torrence AE, Libby RT, Satterfield TF, Lazarowski ER, Gilbert ML, Morton GJ, Bammler TK, Strand AD et al (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  CAS  PubMed  Google Scholar 

  78. Detmer SA, Chan DC (2007) Functions and dysfunctions of mitochondrial dynamics. Nat Rev Mol Cell Biol 8:870–879

    Article  CAS  PubMed  Google Scholar 

  79. Rosenstock TR, Brett AC, Rego AC (2012) Modified mitochondrial dynamics, turnover and function in neurodegeneration: a focus on Huntington’s Disease. Cell Bioenerg Health Dis New Perspect Mitochondrial Biol, 149–194

    Google Scholar 

  80. Shirendeb U, Reddy AP, Manczak M, Calkins MJ, Mao P, Tagle DA, Reddy PH (2011) Abnormal mitochondrial dynamics, mitochondrial loss and mutant huntingtin oligomers in Huntington’s disease: implications for selective neuronal damage. Hum Mol Genet 20:1438–1455

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Reddy PH, Shirendeb UP (2012) Mutant huntingtin, abnormal mitochondrial dynamics, defective axonal transport of mitochondria, and selective synaptic degeneration in Huntington’s disease. Biochim Biophys Acta 1822:101–110

    Article  CAS  PubMed  Google Scholar 

  82. Knott AB, Perkins G, Schwarzenbacher R, Bossy-Wetzel E (2008) Mitochondrial fragmentation in neurodegeneration. Nat Rev Neurosci 9:505–518

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Loson OC, Song Z, Chen H, Chan DC (2013) Fis1, Mff, MiD49, and MiD51 mediate Drp1 recruitment in mitochondrial fission. Mol Biol Cell 24(5):659–667

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Griffin EE, Detmer SA, Chan DC (2006) Molecular mechanism of mitochondrial membrane fusion. Biochim Biophys Acta 1763:482–489

    Article  CAS  PubMed  Google Scholar 

  85. Costa V, Giacomello M, Hudec R, Lopreiato R, Ermak G, Lim D, Malorni W, Davies KJ, Carafoli E, Scorrano L (2010) Mitochondrial fission and cristae disruption increase the response of cell models of Huntington’s disease to apoptotic stimuli. EMBO Mol Med 2:490–503

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Zinsmaier KE, Babic M, Russo GJ (2009) Mitochondrial transport dynamics in axons and dendrites. Results Probl Cell Differ 48:107–139

    CAS  PubMed  Google Scholar 

  87. Lin MY, Sheng ZH (2015) Regulation of mitochondrial transport in neurons. Exp Cell Res 334:35–44

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. 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 Off J Soc Neurosci 28:2783–2792

    Article  CAS  Google Scholar 

  89. Trushina E, Dyer RB, Badger JD 2nd, Ure D, Eide L, Tran DD, Vrieze BT, Legendre-Guillemin V, McPherson PS, Mandavilli BS et al (2004) Mutant huntingtin impairs axonal trafficking in mammalian neurons in vivo and in vitro. Mol Cell Biol 24:8195–8209

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Chang TWD, Rintoul GL, Pandipati S, Reynolds IJ (2006) Mutant huntingtin aggregates impair mitochondrial movement and trafficking in cortical neurons. Neurobiol Dis 22(2):388–400

    Article  CAS  PubMed  Google Scholar 

  91. Twig G, Shirihai OS (2011) The interplay between mitochondrial dynamics and mitophagy. Antioxid Redox Signal 14:1939–1951

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Allen GF, Toth R, James J, Ganley IG (2013) Loss of iron triggers PINK1/Parkin-independent mitophagy. EMBO Rep 14:1127–1135

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Strappazzon F, Nazio F, Corrado M, Cianfanelli V, Romagnoli A, Fimia GM, Campello S, Nardacci R, Piacentini M, Campanella M et al (2015) AMBRA1 is able to induce mitophagy via LC3 binding, regardless of PARKIN and p62/SQSTM1. Cell Death Differ 22:517

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Chu CT, Ji J, Dagda RK, Jiang JF, Tyurina YY, Kapralov AA, Tyurin VA, Yanamala N, Shrivastava IH, Mohammadyani D et al (2013) Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nat Cell Biol 15:1197–1205

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Deas E, Plun-Favreau H, Gandhi S, Desmond H, Kjaer S, Loh SH, Renton AE, Harvey RJ, Whitworth AJ, Martins LM et al (2011) PINK1 cleavage at position A103 by the mitochondrial protease PARL. Hum Mol Genet 20:867–879

    Article  CAS  PubMed  Google Scholar 

  96. Greene AW, Grenier K, Aguileta MA, Muise S, Farazifard R, Haque ME, McBride HM, Park DS, Fon EA (2012) Mitochondrial processing peptidase regulates PINK1 processing, import and Parkin recruitment. EMBO Rep 13:378–385

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Okatsu K, Kimura M, Oka T, Tanaka K, Matsuda N (2015) Unconventional PINK1 localization to the outer membrane of depolarized mitochondria drives Parkin recruitment. J Cell Sci 128:964–978

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Kazlauskaite A, Martinez-Torres RJ, Wilkie S, Kumar A, Peltier J, Gonzalez A, Johnson C, Zhang J, Hope AG, Peggie M et al (2015) Binding to serine 65-phosphorylated ubiquitin primes Parkin for optimal PINK1-dependent phosphorylation and activation. EMBO Rep 16:939–954

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Narendra D, Tanaka A, Suen DF, Youle RJ (2008) Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J Cell Biol 183:795–803

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Khalil B, El Fissi N, Aouane A, Cabirol-Pol MJ, Rival T, Lievens JC (2015) PINK1-induced mitophagy promotes neuroprotection in Huntington’s disease. Cell Death Dis 6:e1617

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Wong YC, Holzbaur EL (2014) The regulation of autophagosome dynamics by huntingtin and HAP1 is disrupted by expression of mutant huntingtin, leading to defective cargo degradation. J Neurosci Off J Soc Neurosci 34:1293–1305

    Article  CAS  Google Scholar 

  102. Rui YN, Xu Z, Patel B, Chen Z, Chen D, Tito A, David G, Sun Y, Stimming EF, Bellen HJ et al (2015) Huntingtin functions as a scaffold for selective macroautophagy. Nat Cell Biol 17:262–275

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Sun N, Yun J, Liu J, Malide D, Liu C, Rovira II, Holmstrom KM, Fergusson MM, Yoo YH, Combs CA et al (2015) Measuring In Vivo Mitophagy. Mol Cell 60:685–696

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Kuwana T, Newmeyer DD (2003) Bcl-2-family proteins and the role of mitochondria in apoptosis. Curr Opin Cell Biol 15:691–699

    Article  CAS  PubMed  Google Scholar 

  105. Parsons MJ, Green DR (2010) Mitochondria in cell death. Essays Biochem 47:99–114

    Article  CAS  PubMed  Google Scholar 

  106. Tait SW, Green DR (2012) Mitochondria and cell signalling. J Cell Sci 125:807–815

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Culmsee C, Mattson MP (2005) p53 in neuronal apoptosis. Biochem Biophys Res Commun 331:761–777

    Article  CAS  PubMed  Google Scholar 

  108. Duan W, Peng Q, Masuda N, Ford E, Tryggestad E, Ladenheim B, Zhao M, Cadet JL, Wong J, Ross CA (2008) Sertraline slows disease progression and increases neurogenesis in N171-82Q mouse model of Huntington’s disease. Neurobiol Dis 30:312–322

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Kong PJ, Kil MO, Lee H, Kim SS, Johnson GV, Chun W (2009) Increased expression of Bim contributes to the potentiation of serum deprivation-induced apoptotic cell death in Huntington’s disease knock-in striatal cell line. Neurol Res 31:77–83

    Article  CAS  PubMed  Google Scholar 

  111. Leon R, Bhagavatula N, Ulukpo O, McCollum M, Wei J (2010) BimEL as a possible molecular link between proteasome dysfunction and cell death induced by mutant huntingtin. Eur J Neurosci 31:1915–1925

    Article  PubMed  PubMed Central  Google Scholar 

  112. Sassone J, Colciago C, Marchi P, Ascardi C, Alberti L, Di Pardo A, Zippel R, Sipione S, Silani V, Ciammola A (2010) Mutant Huntingtin induces activation of the Bcl-2/adenovirus E1B 19-kDa interacting protein (BNip3). Cell Death Dis 1:e7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Garcia-Martinez JM, Perez-Navarro E, Xifro X, Canals JM, Diaz-Hernandez M, Trioulier Y, Brouillet E, Lucas JJ, Alberch J (2007) BH3-only proteins Bid and Bim(EL) are differentially involved in neuronal dysfunction in mouse models of Huntington’s disease. J Neurosci Res 85:2756–2769

    Article  CAS  PubMed  Google Scholar 

  114. Hansson O, Castilho RF, Korhonen L, Lindholm D, Bates GP, Brundin P (2001) Partial resistance to malonate-induced striatal cell death in transgenic mouse models of Huntington’s disease is dependent on age and CAG repeat length. J Neurochem 78:694–703

    Article  CAS  PubMed  Google Scholar 

  115. Teles AV, Rosenstock TR, Okuno CS, Lopes GS, Bertoncini CR, Smaili SS (2008) Increase in bax expression and apoptosis are associated in Huntington’s disease progression. Neurosci Lett 438:59–63

    Article  CAS  PubMed  Google Scholar 

  116. Chiang MC, Chen CM, Lee MR, Chen HW, Chen HM, Wu YS, Hung CH, Kang JJ, Chang CP, Chang C et al (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  CAS  PubMed  Google Scholar 

  117. Zhang Y, Ona VO, Li M, Drozda M, Dubois-Dauphin M, Przedborski S, Ferrante RJ, Friedlander RM (2003) Sequential activation of individual caspases, and of alterations in Bcl-2 proapoptotic signals in a mouse model of Huntington’s disease. J Neurochem 87:1184–1192

    Article  CAS  PubMed  Google Scholar 

  118. Goffredo D, Rigamonti D, Zuccato C, Tartari M, Valenza M, Cattaneo E (2005) Prevention of cytosolic IAPs degradation: a potential pharmacological target in Huntington’s Disease. Pharmacol Res 52:140–150

    Article  CAS  PubMed  Google Scholar 

  119. Rosenstock TR, de Brito OM, Lombardi V, Louros S, Ribeiro M, Almeida S, Ferreira IL, Oliveira CR, Rego AC (2011) FK506 ameliorates cell death features in Huntington’s disease striatal cell models. Neurochem Int 59:600–609

    Article  CAS  PubMed  Google Scholar 

  120. Vis JC, Schipper E, de Boer-van Huizen RT, Verbeek MM, de Waal RM, Wesseling P, ten Donkelaar HJ, Kremer B (2005) Expression pattern of apoptosis-related markers in Huntington’s disease. Acta Neuropathol 109:321–328

    Article  CAS  PubMed  Google Scholar 

  121. Ciammola A, Sassone J, Alberti L, Meola G, Mancinelli E, Russo MA, Squitieri F, Silani V (2006) Increased apoptosis, Huntingtin inclusions and altered differentiation in muscle cell cultures from Huntington’s disease subjects. Cell Death Differ 13:2068–2078

    Article  CAS  PubMed  Google Scholar 

  122. Graham RK, Deng Y, Carroll J, Vaid K, Cowan C, Pouladi MA, Metzler M, Bissada N, Wang L, Faull RL et al (2010) Cleavage at the 586 amino acid caspase-6 site in mutant huntingtin influences caspase-6 activation in vivo. J Neurosci 30:15019–15029

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Graham RK, Deng Y, Slow EJ, Haigh B, Bissada N, Lu G, Pearson J, Shehadeh J, Bertram L, Murphy Z et al (2006) Cleavage at the caspase-6 site is required for neuronal dysfunction and degeneration due to mutant huntingtin. Cell 125:1179–1191

    Article  CAS  PubMed  Google Scholar 

  124. Hermel E, Gafni J, Propp SS, Leavitt BR, Wellington CL, Young JE, Hackam AS, Logvinova AV, Peel AL, Chen SF et al (2004) Specific caspase interactions and amplification are involved in selective neuronal vulnerability in Huntington’s disease. Cell Death Differ 11:424–438

    Article  CAS  PubMed  Google Scholar 

  125. Warby SC, Doty CN, Graham RK, Carroll JB, Yang YZ, Singaraja RR, Overall CM, Hayden MR (2008) Activated caspase-6 and caspase-6-cleaved fragments of huntingtin specifically colocalize in the nucleus. Hum Mol Genet 17:2390–2404

    Article  CAS  PubMed  Google Scholar 

  126. Tebbenkamp AT, Green C, Xu G, Denovan-Wright EM, Rising AC, Fromholt SE, Brown HH, Swing D, Mandel RJ, Tessarollo L et al (2011) Transgenic mice expressing caspase-6-derived N-terminal fragments of mutant huntingtin develop neurologic abnormalities with predominant cytoplasmic inclusion pathology composed largely of a smaller proteolytic derivative. Hum Mol Genet 20:2770–2782

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Waldron-Roby E, Ratovitski T, Wang X, Jiang M, Watkin E, Arbez N, Graham RK, Hayden MR, Hou Z, Mori S et al (2012) Transgenic mouse model expressing the caspase 6 fragment of mutant huntingtin. J Neurosci 32:183–193

    Article  CAS  PubMed  Google Scholar 

  128. Gafni J, Papanikolaou T, Degiacomo F, Holcomb J, Chen S, Menalled L, Kudwa A, Fitzpatrick J, Miller S, Ramboz S et al (2012) Caspase-6 activity in a BACHD mouse modulates steady-state levels of mutant huntingtin protein but is not necessary for production of a 586 amino acid proteolytic fragment. J Neurosci 32:7454–7465

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Landles C, Weiss A, Franklin S, Howland D, Bates G (2012) Caspase-6 does not contribute to the proteolysis of mutant huntingtin in the HdhQ150 knock-in mouse model of Huntington’s disease. PLoS Curr 4, e4fd085bfc9973

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors acknowledge financial support from ‘Fundação para a Ciência e a Tecnologia’ (FCT), Portugal (projects ref. EXPL/BIM-MEC/2220/2013 and Pest-C/SAU/LA0001/2013–2014), co-financed by ‘Programa Operacional Temático Factores de Competividade’ (COMPETE) and supported by the European community fund (FEDER). ACR also acknowledges financial support from ‘Santa Casa da Misericórdia de Lisboa’ (SCML)—Mantero Belard Neuroscience Prize 2013, and ‘Fundação Luso-Americana para o Desenvolvimento’ (FLAD)—Life Science 2020, Portugal. LN holds a Ph.D. fellowship from ‘Fundação para a Ciência e a Tecnologia’ (FCT), Portugal (Reference SFRH/BD/86655/2012). CL was supported by ‘Fundação Luso-Americana para o Desenvolvimento’ (FLAD) Life Science 2020 Postdoctoral Fellowship.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to A. Cristina Rego .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer International Publishing AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Carmo, C., Naia, L., Lopes, C., Rego, A.C. (2018). Mitochondrial Dysfunction in Huntington’s Disease. In: Nóbrega, C., Pereira de Almeida, L. (eds) Polyglutamine Disorders. Advances in Experimental Medicine and Biology, vol 1049. Springer, Cham. https://doi.org/10.1007/978-3-319-71779-1_3

Download citation

Publish with us

Policies and ethics