Skip to main content

Mitochondria in Neurodegeneration

  • Chapter
  • First Online:

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 942))

Abstract

Many neurodegenerative diseases demonstrate abnormal mitochondrial morphology and biochemical dysfunction. Alterations are often systemic rather than brain-limited. Mitochondrial dysfunction may arise as a consequence of abnormal mitochondrial DNA, mutated nuclear proteins that interact directly or indirectly with mitochondria, or through unknown causes. In most cases it is unclear where mitochondria sit in relation to the overall disease cascades that ultimately causes neuronal dysfunction and death, and there is still controversy regarding the question of whether mitochondrial dysfunction is a necessary step in neurodegeneration. In this chapter we highlight and catalogue mitochondrial perturbations in some of the major neurodegenerative diseases including Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington’s disease (HD). We consider data that suggest mitochondria may be critically involved in neurodegenerative disease neurodegeneration cascades.

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

Buying options

Chapter
USD   29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD   169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD   219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD   219.99
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

Learn about institutional subscriptions

References

  • Abou-Sleiman PM, Muqit MMK, Wood NW (2006) Expanding insights of mitochondrial dysfunction in Parkinson’s disease. Nat Rev Neurosci 7(3):207–219

    PubMed  CAS  Google Scholar 

  • Ali SF, David SN, Newport GD, Cadet JL, Slikker W Jr (1994) MPTP-induced oxidative stress and neurotoxicity are age-dependent: evidence from measures of reactive oxygen species and striatal dopamine levels. Synapse 18:27–34

    PubMed  CAS  Google Scholar 

  • Anandatheerthavarada HK, Biswas G, Robin M-A, Avadhani NG (2003) Mitochondrial targeting and a novel transmembrane arrest of Alzheimer’s amyloid precursor protein impairs mitochondrial function in neuronal cells. J Cell Biol 161(1):41–54

    PubMed  CAS  Google Scholar 

  • Antuono P, Beyer J (1999) The burden of dementia. A medical and research perspective. Theor Med Bioethics 20(1):3–13

    CAS  Google Scholar 

  • Arenas J, Campos Y, Ribacoba R, Martin MA, Rubio JC, Ablanedo P, Cabello A (1998) Complex I defect in muscle from patients with Huntington’s disease. Ann Neurol 43:397–400

    PubMed  CAS  Google Scholar 

  • Bacman SR, Bradley WG, Moraes CT (2006) Mitochondrial involvement in amyotrophic lateral sclerosis: trigger or target? Mol Neurobiol 33(2):113–131

    PubMed  CAS  Google Scholar 

  • 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

    PubMed  CAS  Google Scholar 

  • Bassett SS, Avramopoulos D, Fallin D (2002) Evidence for parent of origin effect in late-onset Alzheimer disease. Am J Med Genet B Neuropsychiatr Genet 114(6):679–686

    Google Scholar 

  • Beal MF (1995) Aging, energy, and oxidative stress in neurodegenerative diseases. Ann Neurol 38(3):357–366

    PubMed  CAS  Google Scholar 

  • Beal MF (1998) Excitotoxicity and nitric oxide in Parkinson’s disease pathogenesis. Ann Neurol 44:S110–S114

    PubMed  CAS  Google Scholar 

  • 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 13(10):4181–4192

    PubMed  CAS  Google Scholar 

  • Benchoua A, Trioulier Y, Zala D, Gaillard MC, Lefort N, Dufour N, Saudou F, Elalouf JM, Hirsch E, Hantraye P, Déglon 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(4):1652–1663

    PubMed  CAS  Google Scholar 

  • Bender A, Krishnan KJ, Morris CM, Taylor GA, Reeve AK, Perry RH, Jaros E, Hersheson JS, Betts J, Klopstock T, Taylor RW, Turnbull DM (2006) High levels of mitochondrial DNA deletions in substantia nigra neurons in aging and Parkinson disease. Nat Genet 38(5):515–517

    PubMed  CAS  Google Scholar 

  • Betarbet R, Sherer TB, Mackenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT (2000) Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci 3(12):1301–1306

    PubMed  CAS  Google Scholar 

  • Bindoff LA, Birch-Machin M, Cartlidge NEF, Parker WD, Turnbull DM (1989) Mitochondrial function in Parkinson’s disease. Lancet 2(8653):49

    PubMed  CAS  Google Scholar 

  • Borthwick GM, Johnson MA, Ince PG, Shaw PJ, Turnbull DM (1999) Mitochondrial enzyme activity in amyotrophic lateral sclerosis: implications for the role of mitochondria in neuronal cell death. Ann Neurol 46:787–790

    PubMed  CAS  Google Scholar 

  • Bosetti F, Brizzi F, Barogi S, Mancuso M, Siciliano G, Tendi EA, Murri L, Rapoport SI, Solaini G (2002) Cytochrome c oxidase and mitochondrial F1F0-ATPase (ATP synthase) activities in platelets and brain from patients with Alzheimer’s disease. Neurobiol Aging 23(3):371–376

    PubMed  CAS  Google Scholar 

  • Bowling AC, Schulz JB, Brown RH, Beal MF (1993) Superoxide dismutase activity, oxidative damage, and mitochondrial energy metabolism in familial and sporadic amyotrophic lateral sclerosis. J Neurochem 61(6):2322–2325

    PubMed  CAS  Google Scholar 

  • 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

    PubMed  CAS  Google Scholar 

  • 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(5):646–653

    PubMed  CAS  Google Scholar 

  • Browne SE, Bowling AC, Baik MJ, Gurney M, Brown RH, Beal MF (1998) Metabolic dysfunction in familial, but not sporadic, amyotrophic lateral sclerosis. J Neurochem 71:281–287

    PubMed  CAS  Google Scholar 

  • Canevari L, Clark JB, Bates TE (1999) β-Amyloid fragment 25–35 selectively decreases complex IV activity in isolated mitochondria. FEBS Lett 457(1):131–134

    PubMed  CAS  Google Scholar 

  • Cannon JR, Tapias V, Na HM, Honick AS, Drolet RE, Greenamyre JT (2009) A highly reproducible rotenone model of Parkinson’s disease. Neurobiol Dis 34(2):279–290

    PubMed  CAS  Google Scholar 

  • Cardoso SM, Santos S, Swerdlow RH, Oliveira CR (2001) Functional mitochondria are required for amyloid beta-mediated neurotoxicity. FASEB J 15(8):1439–1441

    PubMed  CAS  Google Scholar 

  • Cardoso SM, Proença MT, Santos S, Santana I, Oliveira CR (2004a) Cytochrome c oxidase is decreased in Alzheimer’s disease platelets. Neurobiol Aging 25(1):105–110

    PubMed  CAS  Google Scholar 

  • Cardoso SM, Santana I, Swerdlow RH, Oliveira CR (2004b) Mitochondria dysfunction of Alzheimer’s disease cybrids enhances Abeta toxicity. J Neurochem 89(6):1417–1426

    PubMed  CAS  Google Scholar 

  • Carri MT, Ferri A, Battistoni A, Famhy L, Gabbianelli R, Poccia F, Rotilio G (1997) Expression of a Cu, Zn superoxide dismutase typical of familial amyotrophic lateral sclerosis induces mitochondrial alteration and increase of cytosolic Ca2+ concentration in transfected neuroblastoma SH-SY5Y cells. FEBS Lett 414(2):365–368

    PubMed  CAS  Google Scholar 

  • Casley CS, Canevari L, Land JM, Clark JB, Sharpe MA (2002) β-Amyloid inhibits integrated mitochondrial respiration and key enzyme activities. J Neurochem 80(1):91–100

    PubMed  CAS  Google Scholar 

  • Caspersen C, Wang N, Yao J, Sosunov A, Chen X, Lustbader JW, Xu HW, Stern D, Mckhann G, Yan SD (2005) Mitochondrial Abeta: a potential focal point for neuronal metabolic dysfunction in Alzheimer’s disease. FASEB J 19(14):2040–2041

    PubMed  CAS  Google Scholar 

  • Chang S-W, Zhang D, Chungb HD, Zassenhaus HP (2000) The frequency of point mutations in mitochondrial DNA is elevated in the Alzheimer’s brain. Biochem Biophys Res Commun 273(1):203–208

    PubMed  CAS  Google Scholar 

  • 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(14):1407–1420

    PubMed  CAS  Google Scholar 

  • Chung KKK, Thomas B, Li X, Pletnikova O, Troncoso JC, Marsh L, Dawson VL, Dawson TM (2004) S-nitrosylation of parkin regulates ubiquitination and compromises parkin’s protective function. Science 304(5675):1328–1331

    PubMed  CAS  Google Scholar 

  • Corral-Debrinski M, Horton T, Lott MT, Shoffner JM, Mckee AC, Beal MF, Graham BH, Wallace DC (1994) Marked changes in mitochondrial DNA deletion levels in Alzheimer brains. Genomics 23:471–476

    PubMed  CAS  Google Scholar 

  • Crouch PJ, Blake R, Duce JA, Ciccotosto GD, Li Q-X, Barnham KJ, Curtain CC, Cherny RA, Cappai R, Dyrks T, Masters CL, Trounce IA (2005) Copper-dependent inhibition of human cytochrome c oxidase by a dimeric conformer of amyloid-beta1-42. J Neurosci 25(3):672–679

    PubMed  CAS  Google Scholar 

  • 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(1):59–69

    PubMed  CAS  Google Scholar 

  • Curti D, Rognoni F, Gasparini L, Cattaneo A, Paolillo M, Racchi M, Zani L, Bianchetti A, Trabucchi M, Bergamaschi S, Govoni S (1997) Oxidative metabolism in cultured fibroblasts derived from sporadic Alzheimer’s disease (AD) patients. Neurosci Lett 236(1):13–16

    PubMed  CAS  Google Scholar 

  • Damiano M, Starkov AA, Petri S, Kipiani K, Kiaei M, Mattiazzi M, Beal MF, Manfredi G (2006) Neural mitochondrial Ca2+ capacity impairment precedes the onset of motor symptoms in G93A Cu/Zn-superoxide dismutase mutant mice. J Neurochem 96(5):1349–1361

    PubMed  CAS  Google Scholar 

  • De La Monte SM, Luong T, Neely TR, Robinson D, Wands JR (2000) Mitochondrial DNA damage as a mechanism of cell loss in Alzheimer’s disease. Lab Invest 80(8):1323–1335

    PubMed  Google Scholar 

  • Devi L, Prabhu BM, Galati DF, Avadhani NG, Anandatheerthavarada HK (2006) Accumulation of amyloid precursor protein in the mitochondrial import channels of human Alzheimer’s disease brain is associated with mitochondrial dysfunction. J Neurosci 26(35):9057–9068

    PubMed  CAS  Google Scholar 

  • Dhaliwal GK, Grewal RP (2000) Mitochondrial DNA deletion mutation levels are elevated in ALS brains. Neuroreport 11(11):2507–2509

    PubMed  CAS  Google Scholar 

  • Diana A, Simic G, Sinforiani E, Orru N, Pichiri G, Bono G (2008) Mitochondria morphology and DNA content upon sublethal exposure to beta-amyloid(1-42) peptide. Coll Antropol 32(Suppl 1):51–58

    PubMed  CAS  Google Scholar 

  • Duara R, Lopez-Alberola RF, Barker WW, Loewenstein DA, Zatinsky M, Eisdorfer CE, Weinberg GB (1993) A comparison of familial and sporadic Alzheimer’s disease. Neurology 43:1377–1384

    PubMed  CAS  Google Scholar 

  • Edland SD, Silverman JM, Peskind ER, Tsuang D, Wijsman E, Morns JC (1996) Increased risk of dementia in mothers of Alzheimer’s disease cases: evidence for maternal inheritance. Neurology 47:254–256

    PubMed  CAS  Google Scholar 

  • Ehrenkrantz D, Silverman JM, Smith CJ, Birstein S, Marin D, Mohs RC, Davis KL (1999) Genetic epidemiological study of maternal and paternal transmission of Alzheimer’s disease. Am J Med Genet B Neuropsychiatr Genet 88:378–382

    CAS  Google Scholar 

  • Elson JL, Herrnstadt C, Preston G, Thal L, Morris CM, Edwardson JA, Beal MF, Turnbull DM, Howell N (2006) Does the mitochondrial genome play a role in the etiology of Alzheimer’s disease? Hum Genet 119:241–254

    PubMed  CAS  Google Scholar 

  • Esteves ARF, Domingues F, Ferreira IL, Januário C, Swerdlow RH, Oliveira CR, Cardoso SM (2008) Mitochondrial function in Parkinson’s disease cybrids containing an nt2 neuron-like nuclear background. Mitochondrion 8(3):219–228

    PubMed  CAS  Google Scholar 

  • Esteves AR, Lu J, Rodova M, Onyango I, Lezi E, Dubinsky R, Lyons KE, Pahwa R, Burns JM, Cardoso SM, Swerdlow RH (2010) Mitochondrial respiration and respiration associated proteins in cell lines created through Parkinson’s subject mitochondrial transfer. J Neurochem 113(3):674–682

    PubMed  CAS  Google Scholar 

  • Fujita K, Yamauchi M, Shibayama K, Ando M, Honda M, Nagata Y (1996) Decreased cytochrome c oxidase activity but unchanged superoxide dismutase and glutathione peroxidase activities in the spinal cords of patients with amyotrophic lateral sclerosis. J Neurosci Res 45:276–281

    PubMed  CAS  Google Scholar 

  • Gabbita SP, Lovell MA, Markesbery WR (1998) Increased nuclear DNA oxidation in the brain in Alzheimer’s disease. J Neurochem 71:2034–2040

    PubMed  CAS  Google Scholar 

  • 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

    PubMed  CAS  Google Scholar 

  • Gu M, Cooper JM, Taanman JW, Schapira AHV (1998) Mitochondrial DNA transmission of the mitochondrial defect in Parkinson’s disease. Ann Neurol 44:177–186

    PubMed  CAS  Google Scholar 

  • Hambleta NS, Castora FJ (1997) Elevated levels of the Kearns-Sayre syndrome mitochondrial DNA deletion in temporal cortex of Alzheimer’s patients. Mutat Res 379:253–262

    Google Scholar 

  • Hansson CA, Frykman S, Farmery MR, Tjernberg LO, Nilsberth C, Pursglove SE, Ito A, Winblad B, Cowburn RF, Thyberg J, Ankarcrona M (2004) Nicastrin, presenilin, APH-1, and PEN-2 form active gamma-secretase complexes in mitochondria. J Biol Chem 279:51654–51660

    PubMed  CAS  Google Scholar 

  • Hantraye P, Brouillet E, Ferrante R, Palfi S, Dolan R, Matthews RT, Beal MF (1996) Inhibition of neuronal nitric oxide synthase prevents MPTP-induced parkinsonism in baboons. Nat Med 2:1017–1021

    PubMed  CAS  Google Scholar 

  • Hardy JA, Higgins GA (1992) Alzheimer’s disease: the amyloid cascade hypothesis. Science 256:184–185

    PubMed  CAS  Google Scholar 

  • Hattingen E, Magerkurth J, Pilatus U, Mozer A, Seifried C, Steinmetz H, Zanella F, Hilker R (2009) Phosphorus and proton magnetic resonance spectroscopy demonstrates mitochondrial dysfunction in early and advanced Parkinson’s disease. Brain 132:3285–3297

    PubMed  Google Scholar 

  • Henchcliffe C, Shungu DC, Mao X, Huang C, Nirenberg MJ, Jenkins BG, Beal MF (2008) Multinuclear magnetic resonance spectroscopy for in vivo assessment of mitochondrial dysfunction in Parkinson’s disease. Ann N Y Acad Sci 1147:206–220

    PubMed  CAS  Google Scholar 

  • Higgins CMJ, Jung C, Ding H, Xu Z (2002) Mutant Cu, Zn superoxide dismutase that causes motoneuron degeneration is present in mitochondria in the CNS. J Neurosci 22:RC215

    PubMed  Google Scholar 

  • Higgins CM, Jung C, Xu Z (2003) ALS-associated mutant SOD1G93A causes mitochondrial vacuolation by expansion of the intermembrane space and by involvement of SOD1 aggregation and peroxisomes. BMC Neurosci 4:16

    PubMed  Google Scholar 

  • Hirai K, Aliev G, Nunomura A, Fujioka H, Russell RL, Atwood CS, Johnson AB, Kress Y, Vinters HV, Tabaton M, Shimohama S, Cash AD, Siedlak SL, Harris PLR, Jones PK, Petersen RB, Perry G, Smith MA (2001) Mitochondrial abnormalities in Alzheimer’s disease. J Neurosci 21:3017–3023

    PubMed  CAS  Google Scholar 

  • Hirano A, Donnenfeld H, Sasaki S, Nakano I (1984) Fine structural observations of neuroWlamentous changes in amyotrophic lateral sclerosis. J Neuropathol Exp Neurol 43:461–470

    PubMed  CAS  Google Scholar 

  • 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

    Google Scholar 

  • Ikebe S, Tanaka M, Ohno K, Sato W, Hattori K, Kondo T, Mizuno Y, Ozawa T (1990) Increase of deleted mitochondrial DNA in the striatum in Parkinson’s disease and senescence. Biochem Biophys Res Commun 170:1044–1048

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

  • Jung C, Higgins CMJ, Xu Z (2002) A quantitative histochemical assay for activities of mitochondrial electron transport chain complexes in mouse spinal cord sections. J Neurosci Methods 114:165–172

    PubMed  CAS  Google Scholar 

  • Kabashi E, Valdmanis PN, Dion P, Spiegelman D, Mcconkey BJ, Vande Velde C, Bouchard JP, Lacomblez L, Pochigaeva K, Salachas F, Pradat PF, Camu W, Meininger V, Dupre N, Rouleau GA (2008) TARDBP mutations in individuals with sporadic and familial amyotrophic lateral sclerosis. Nat Genet 40:572–574

    PubMed  CAS  Google Scholar 

  • Khan SM, Cassarino DS, Abramova NN, Keeney PM, Borland MK, Trimmer PA, Krebs CT, Bennett JC, Parks JK, Swerdlow RH, Parker WD, Bennett JP (2000) Alzheimer’s disease cybrids replicate beta-amyloid abnormalities through cell death pathways. Ann Neurol 48:148–155

    PubMed  CAS  Google Scholar 

  • Kish SJ, Bergeron C, Rajput A, Dozic S, Mastrogiacomo F, Chang LJ, Wilson JM, Distefano LM, Nobrega JN (1992) Brain cytochrome oxidase in Alzheimer’s disease. J Neurochem 59:776–779

    PubMed  CAS  Google Scholar 

  • Kong J, Xu Z (1998) Massive mitochondrial degeneration in motor neurons triggers the onset of amyotrophic lateral sclerosis in mice expressing a mutant SOD1. J Neurosci 18:3241–3250

    PubMed  CAS  Google Scholar 

  • Kraytsberg Y, Kudryavtseva E, Mckee AC, Geula C, Kowall NW, Khrapko K (2006) Mitochondrial DNA deletions are abundant and cause functional impairment in aged human substantia nigra neurons. Nat Genet 38:518–520

    PubMed  CAS  Google Scholar 

  • Kuhl DE, Phelps ME, Markham CH, Metter EJ, Riege WH, Winter J (1982) Cerebral metabolism and atrophy in Huntington’s disease determined by 18FDG and computed tomographic scan. Ann Neurol 12:425–434

    PubMed  CAS  Google Scholar 

  • Kwiatkowski TJ, Bosco DA, Leclerc AL, Tamrazian E, Vanderburg CR, Russ C, Davis A, Gilchrist J, Kasarskis EJ, Munsat T, Valdmanis P, Rouleau GA, Hosler BA, Cortelli P, De Jong PJ, Yoshinaga Y, Haines JL, Pericak-Vance MA, Yan J, Ticozzi N, Siddique T, Mckenna-Yasek D, Sapp PC, Horvitz HR, Landers JE, Brown RH (2009) Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science 323:1205–1208

    PubMed  CAS  Google Scholar 

  • Lestienne P, Nelson I, Riederer P, Reichmann H, Jellinger K (1991) Mitochondrial DNA in postmortem brain from patients with Parkinson’s disease. J Neurochem 56:1819

    PubMed  CAS  Google Scholar 

  • Lin J, Wu PH, Tarr PT, Lindenberg KS, St-Pierre J, Zhang CY, Mootha VK, Jäger 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

    PubMed  CAS  Google Scholar 

  • Liu J, Lillo C, Jonsson PA, Velde CV, Ward CM, Miller TM, Subramaniam JR, Rothstein JD, Marklund S, Andersen PM, Brännström T, Gredal O, Wong PC, Williams DS, Cleveland DW (2004) Toxicity of familial ALS-linked SOD1 mutants from selective recruitment to spinal mitochondria. Neuron 43:5–17

    PubMed  CAS  Google Scholar 

  • Lustbader JW, Cirilli M, Lin C, Xu HW, Takuma K, Wang N, Caspersen C, Chen X, Pollak S, Chaney M, Trinchese F, Liu S, Gunn-Moore F, Lue L-F, Walker DG, Kuppusamy P, Zewier ZL, Arancio O, Stern D, Yan SS, Wu H (2004) ABAD directly links Abeta to mitochondrial toxicity in Alzheimer’s disease. Science 304:448–452

    PubMed  CAS  Google Scholar 

  • Mancuso M, Conforti FL, Rocchi A, Tessitore A, Muglia M, Tedeschi G, Panza D, Monsurrò M, Sola P, Mandrioli J, Choub A, Delcorona A, Manca ML, Mazzei R, Sprovieri T, Filosto M, Salviati A, Valentino P, Bono F, Caracciolo M, Simone IL, Bella VL, Majorana G, Siciliano G, Murri L, Quattrone A (2004) Could mitochondrial haplogroups play a role in sporadic amyotrophic lateral sclerosis? Neurosci Lett 371:158–162

    PubMed  CAS  Google Scholar 

  • Manczak M, Anekonda TS, Henson E, Park BS, Quinn J, Reddy PH (2006) Mitochondria are a direct site of A beta accumulation in Alzheimer’s disease neurons: implications for free radical generation and oxidative damage in disease progression. Hum Mol Genet 15:1437–1449

    PubMed  CAS  Google Scholar 

  • Martin LJ, Pan Y, Price AC, Sterling W, Copeland NG, Jenkins NA, Price DL, Lee MK (2006) Parkinson’s disease alpha-synuclein transgenic mice develop neuronal mitochondrial degeneration and cell death. J Neurosci 26:41–50

    PubMed  CAS  Google Scholar 

  • Masui Y, Mozai T, Kakehi K (1985) Functional and morphometric study of the liver in motor neuron disease. J Neurol 232:15–19

    PubMed  CAS  Google Scholar 

  • Mecocci P, Macgarvey U, Beal MF (1994) Oxidative damage to mitochondrial DNA is increased in Alzheimer’s disease. Ann Neurol 36(5):747–751

    PubMed  CAS  Google Scholar 

  • Menzies FM, Cookson MR, Taylor RW, Turnbull DM, Chrzanowska-Lightowlers ZMA, Dong L, Figlewicz DA, Shaw PJ (2002) Mitochondrial dysfunction in a cell culture model of familial amyotrophic lateral sclerosis. Brain 125:1522–1533

    PubMed  Google Scholar 

  • Mizuno Y, Suzuki K, Sone N, Saitoh T (1988) Inhibition of mitochondrial respiration by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in mouse brain in vivo. Neurosci Lett 91:349–353

    PubMed  CAS  Google Scholar 

  • Mosconi L (2005) Brain glucose metabolism in the early and specific diagnosis of Alzheimer’s disease. FDG-PET studies in MCI and AD. Eur J Nucl Med Mol Imaging 32(4):486–510

    PubMed  CAS  Google Scholar 

  • Mosconi L, Brys M, Switalski R, Mistur R, Glodzik L, Pirraglia E, Tsui W, Santi SD, Leon MJD (2007) Maternal family history of Alzheimer’s disease predisposes to reduced brain glucose metabolism. Proc Natl Acad Sci USA 104(48):19067–19072

    PubMed  CAS  Google Scholar 

  • Mosconi L, Mistur R, Switalski R, Brys M, Glodzik L, Rich K, Pirraglia E, Tsui W, Santi SD, Leon MJD (2009) Declining brain glucose metabolism in normal individuals with a maternal history of Alzheimer disease. Neurology 72:513–520

    PubMed  CAS  Google Scholar 

  • Murata T, Ohtsuka C, Terayama Y (2008) Increased mitochondrial oxidative damage and oxidative DNA damage contributes to the neurodegenerative process in sporadic amyotrophic lateral sclerosis. Free Radic Res 42:221–225

    PubMed  CAS  Google Scholar 

  • Mutisya EM, Bowling AC, Beal MF (1994) Cortical cytochrome oxidase activity is reduced in Alzheimer’s disease. J Neurochem 63(6):2179–2184

    PubMed  CAS  Google Scholar 

  • Mytilineou C, Werner P, Molinari S, Di-Rocco A, Cohen G, Yahr MD (1994) Impaired oxidative decarboxylation of pyruvate in fibroblasts from patients with Parkinson’s disease. J Neural Transm Parkinson’s Dis Dement Sect 8(3):223–228

    CAS  Google Scholar 

  • Nakano Y, Hirayama K, Terao K (1987) Hepatic ultrastructural changes and liver dysfunction in amyotrophic lateral sclerosis. Arch Neurol 44:103–106

    PubMed  CAS  Google Scholar 

  • 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

    PubMed  CAS  Google Scholar 

  • Ng MC, Iacopino AM, Quintero EM, Marches F, Sonsalla PK, Liang CL, Speciale SG, German DC (1996) The neurotoxin MPTP increases calbindin-D28k levels in mouse midbrain dopaminergic neurons. Brain Res Mol Brain Res 36:329–336

    PubMed  CAS  Google Scholar 

  • Nicklas WJ, Vyas I, Heikkila RE (1985) Inhibition of NADH-linked oxidation in brain mitochondria by 1-methyl-4-phenyl-pyridine, a metabolite of the neurotoxin, 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine. Life Sci 36:2503–2508

    PubMed  CAS  Google Scholar 

  • Palacino JJ, Sagi D, Goldberg MS, Krauss S, Motz C, Wacker M, Klose J, Shen J (2004) Mitochondrial dysfunction and oxidative damage in parkin-deficient mice. J Biol Chem 279:18614–18622

    PubMed  CAS  Google Scholar 

  • 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 

  • Parker WD, Boyson SJ, Parks JK (1989) Abnormalities of the electron transport chain in idiopathic Parkinson’s disease. Ann Neurol 26:719–723

    PubMed  Google Scholar 

  • Parker WD, Filley CM, Parks JK (1990a) Cytochrome oxidase deficiency in Alzheimer’s disease. Neurology 40:1302–1303

    PubMed  Google Scholar 

  • Parker WD Jr, Boyson SJ, Luder AS, Parks JK (1990b) Evidence for a defect in NADH: ubiquinone oxidoreductase (complex I) in Huntington’s disease. Neurology 40:1231–1234

    PubMed  Google Scholar 

  • Parker WD, Mahr NJ, Filley CM, Parks JK, Hughes D, Young DA, Cullum CM (1994) Reduced platelet cytochrome c oxidase activity in Alzheimer’s disease. Neurology 44:1086–1090

    PubMed  Google Scholar 

  • Parker JA, Arango M, Abderrahmane S, Lambert E, Tourette C, Catoire H, Néri C (2005) Resveratrol rescues mutant polyglutamine cytotoxicity in nematode and mammalian neurons. Nat Genet 37:349–350

    PubMed  CAS  Google Scholar 

  • Parker WD, Parksa JK, Swerdlow RH (2008) Complex I deficiency in Parkinson’s disease frontal cortex. Brain Res 1189:215–218

    PubMed  CAS  Google Scholar 

  • Payami H, Hoffbuhr K (1993) Lack of evidence for maternal effect in familial Alzheimer’s disease. Genet Epidemiol 10:461–464

    PubMed  CAS  Google Scholar 

  • Pereira C, Santos MS, Oliveira C (1998) Mitochondrial function impairment induced by amyloid peptide on PC12 cells. Neuroreport 9:1749–1755

    CAS  Google Scholar 

  • Pesah Y, Pham T, Burgess H, Middlebrooks B, Verstreken P, Zhou Y, Harding M, Bellen H, Mardon G (2004) Drosophila parkin mutants have decreased mass and cell size and increased sensitivity to oxygen radical stress. Development 131:2183–2194

    PubMed  CAS  Google Scholar 

  • Petit A, Kawarai T, Paitel E, Sanjo N, Maj M, Scheid M, Chen F, Gu Y, Hasegawa H, Salehi-Rad S, Wang L, Rogaeva E, Fraser P, Robinson B, George-Hyslop PS, Tandon A (2005) Wild-type PINK1 prevents basal and induced neuronal apoptosis, a protective effect abrogated by Parkinson disease-related mutations. J Biol Chem 280:34025–34032

    PubMed  CAS  Google Scholar 

  • Praticò D, Uryu K, Leight S, Trojanoswk JQ, Lee VM-Y (2001) Increased lipid peroxidation precedes amyloid plaque formation in an animal model of Alzheimer amyloidosis. J Neurosci 21:4183–4187

    PubMed  Google Scholar 

  • Raimondi A, Mangolini A, Rizzardini M, Tartari S, Massari S, Bendotti C, Francolini M, Borgese N, Cantoni L, Pietrini G (2006) Cell culture models to investigate the selective vulnerability of motoneuronal mitochondria to familial ALS-linked G93ASOD1. Eur J Neurosci 24:387–399

    PubMed  Google Scholar 

  • Ro L-S, Lai S-L, Chen C-M, Chen S-T (2003) Deleted 4977-bp mitochondrial DNA mutation is associated with sporadic amyotrophic lateral sclerosis: a hospital-based case-control study. Muscle Nerve 28:737–743

    PubMed  CAS  Google Scholar 

  • Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P (2005) Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature 434:113–118

    PubMed  CAS  Google Scholar 

  • Rui Y, Tiwari P, Xie Z, Zheng JQ (2006) Acute Impairment of Mitochondrial Trafficking by beta–Amyloid Peptides in Hippocampal Neurons. J Neurosci 26:10480–10487

    PubMed  CAS  Google Scholar 

  • Sanberg PR, Fibiger HC, Mark RF (1981) Body weight and dietary factors in Huntington’s disease patients compared with matched controls. Med J Aust 1:407–409

    PubMed  CAS  Google Scholar 

  • Sasaki S, Iwata M (1996) Impairment of fast axonal transport in the proximal axons of anterior horn neurons in amyotrophic lateral sclerosis. Neurology 47:535–540

    PubMed  CAS  Google Scholar 

  • Schapira AHV, Cooper JM, Dexter D, Jenner P, Clark JB, Marsden CD (1989) Mitochondrial complex I deficiency in Parkinson’s disease. Lancet 1:1269

    PubMed  CAS  Google Scholar 

  • Schapira AHV, Holt IJ, Sweeney M, Harding AE, Jenner P, Marsden CD (1990) Mitochondrial DNA analysis in Parkinson’s disease. Mov Disord 5(4):294–297

    PubMed  CAS  Google Scholar 

  • Scheuner D, Eckman C, Jensen M, Song X, Citron M, Suzuki N, Bird T, Hardy J, Hutton M, Kukull W, Larson E, Levy-Lahad E, Viitanen M, Peskind E, Poorkaj P, Schellenberg G, Tanzi R, Wasco W, Lannfelt L, Selkoe D, Younkin S (1996) Secreted amyloid beta-protein similar to that in the senile plaques of Alzheimer’s disease is increased in vivo by the presenilin 1 and 2 and APP mutations linked to familial Alzheimer’s disease. Nat Med 2:864–870

    PubMed  CAS  Google Scholar 

  • Sheehan JP, Swerdlow RH, Parker WD, Miller SW, Davis RE, Tuttle JB (1997) Altered calcium homeostasis in cells transformed by mitochondria from individuals with Parkinson’s disease. J Neurochem 68:1221–1233

    PubMed  CAS  Google Scholar 

  • Shibata N, Nagai R, Uchida K, Horiuchi S, Yamada S, Hirano A, Kawaguchi M, Yamamoto T, Sasaki S, Kobayashi M (2001) Morphological evidence for lipid peroxidation and protein glycoxidation in spinal cords from sporadic amyotrophic lateral sclerosis patients. Brain Res 917:97–104

    PubMed  CAS  Google Scholar 

  • Siciliano G, D’avino C, Del Corona A, Barsacchi R, Kusmic C, Rocchi A, Pastorini E, Murri L (2002) Impaired oxidative metabolism and lipid peroxidation in exercising muscle from ALS patients. Amyotroph Lateral Scler Other Motor Neuron Disord 3:57–62

    PubMed  CAS  Google Scholar 

  • Silvestri L, Caputo V, Bellacchio E, Atorino L, Dallapiccola B, Valente EM, Casari G (2005) Mitochondrial import and enzymatic activity of PINK1 mutants associated to recessive parkinsonism. Hum Mol Genet 14:3477–3492

    PubMed  CAS  Google Scholar 

  • Simpson EP, Henry YK, Henkel JS, Smith RG, Appel SH (2004) Increased lipid peroxidation in sera of ALS patients: a potential biomarker of disease burden. Neurology 62:1758–1765

    PubMed  CAS  Google Scholar 

  • Smith TS, Swerdlow RH, Parker WD Jr, Bennett JP Jr (1994) Reduction of MPP(+)-induced hydroxyl radical formation and nigrostriatal MPTP toxicity by inhibiting nitric oxide synthase. Neuroreport 5:2598–2600

    PubMed  CAS  Google Scholar 

  • Song DD, Shults CW, Sisk A, Rockenstein E, Masliah E (2004) Enhanced substantia nigra mitochondrial pathology in human alpha-synuclein transgenic mice after treatment with MPTP. Exp Neurol 186:158–172

    PubMed  CAS  Google Scholar 

  • Spillantini MG, Schmidt ML, Lee VM-Y, Trojanowski JQ, Jakes R, Goedert M (1997) Alpha-synuclein in Lewy bodies. Nature 388:839–840

    PubMed  CAS  Google Scholar 

  • Sreedharan J, Blair IP, Tripathi VB, Hu X, Vance C, Rogelj B, Ackerley S, Durnall JC, Williams KL, Buratti E, Baralle F, De Belleroche J, Mitchell JD, Leigh PN, Al-Chalabi A, Miller CC, Nicholson G, Shaw CE (2008) TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science 319:str. 1668–str. 1672

    Google Scholar 

  • Sugars KL, Rubinsztein DC (2003) Transcriptional abnormalities in Huntington disease. Trends Genet 19:233–238

    PubMed  CAS  Google Scholar 

  • Swerdlow RH (2000) Role of mitochondria in Parkinson’s disease. In: Molecular mechanisms of neurodegenerative diseases. Humana Press Inc, Totowa

    Google Scholar 

  • Swerdlow RH (2007a) Is aging part of Alzheimer’s disease, or is Alzheimer’s disease part of aging? Neurobiol Aging 28:1465–1480

    PubMed  Google Scholar 

  • Swerdlow RH (2007b) Mitochondria in cybrids containing mtDNA from persons with mitochondriopathies. J Neurosci Res 85:3416–3428

    PubMed  CAS  Google Scholar 

  • Swerdlow RH (2007c) Pathogenesis of Alzheimer’s disease. Clin Interv Aging 2:347–359

    PubMed  CAS  Google Scholar 

  • Swerdlow RH (2009) The neurodegenerative mitochondriopathies. J Alzheimers Dis 17:737–751

    PubMed  CAS  Google Scholar 

  • Swerdlow RH, Khan SM (2004) A “mitochondrial cascade hypothesis” for sporadic Alzheimer’s disease. Med Hypotheses 63:8–20

    PubMed  CAS  Google Scholar 

  • Swerdlow RH, Khan SM (2009) The Alzheimer’s disease mitochondrial cascade hypothesis: an update. Exp Neurol 218:308–315

    PubMed  CAS  Google Scholar 

  • Swerdlow RH, Kish SJ (2002) Mitochondria in Alzheimer’s disease. Int Rev Neurobiol 53:341–385

    PubMed  CAS  Google Scholar 

  • Swerdlow RH, Parks JK, Miller SW, Davis RE, Tuttle JB, Trimmer PA, Sheehan JP, Bennett JP, Parker WD (1996a) Origin and functional consequences of the complex I defect in Parkinson’s disease. Ann Neurol 40:663–671

    PubMed  CAS  Google Scholar 

  • Swerdlow RH, Parks JK, Miller SW, Pattee G, Davis RE, Parker WD (1996b) Evidence of genetic mitochondrial pathology in sporadic Amyotrophic Lateral Sclerosis. Soc Neurosci Abst 22:94.5

    Google Scholar 

  • Swerdlow RH, Parks JK, Cassarino DS, Maguire DJ, Maguire RS, Bennett JP, Davis RE, Parker WD (1997) Cybrids in Alzheimer’s disease: a cellular model of the disease? Neurology 49:918–925

    PubMed  CAS  Google Scholar 

  • Swerdlow RH, Parks JK, Cassarino DS, Trimmer PA, Miller SW, Maguire DJ, Sheehan JP, Maguire RS, Pattee G, Juel VC, Phillips LH, Tuttle JB, Bennett JP, Davis RE, Parker WD (1998) Mitochondria in sporadic amyotrophic lateral sclerosis. Exp Neurol 153:135–142

    PubMed  CAS  Google Scholar 

  • Swerdlow RH, Parks JK, Pattee G, Parker WD (2000) Role of mitochondria in amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Motor Neuron Disord 1:185–190

    PubMed  CAS  Google Scholar 

  • Swerdlow RH, Parker WD, Currie LJ, Bennett JP, Harrison MB, Trugman JM, Wooten GF (2001) Gender ratio differences between Parkinson’s disease patients and their affected parents. Parkinsonism Relat Disord 7:129–133

    PubMed  Google Scholar 

  • Tanner CM, Goldman SM (1996) Epidemiology of Parkinson’s disease. Neurol Clin 14:317–335

    PubMed  CAS  Google Scholar 

  • Trimmer PA, Bennett JP (2009) The cybrid model of sporadic Parkinson’s disease. Exp Neurol 218:320–325

    PubMed  CAS  Google Scholar 

  • Trimmer PA, Borland MK, Keeney PM, Bennett JP, Parker WD (2004) Parkinson’s disease transgenic mitochondrial cybrids generate Lewy inclusion bodies. J Neurochem 88:800–812

    PubMed  CAS  Google Scholar 

  • Valavanidis A, Vlachogianni T, Fiotakis C (2009) 8-hydroxy-2′ -deoxyguanosine (8-OHdG): a critical biomarker of oxidative stress and carcinogenesis. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev 27:20–39

    Google Scholar 

  • Valla J, Schneider L, Niedzielko T, Coon KD, Caselli R, Sabbagh MN, Ahern GL, Baxter L, Alexander G, Walker DG, Reiman EM (2006) Impaired platelet mitochondrial activity in Alzheimer’s disease and mild cognitive impairment. Mitochondrion 6:323–330

    PubMed  CAS  Google Scholar 

  • Van Der Walt JM, Nicodemus KK, Martin ER, Scott WK, Nance MA, Watts RL, Hubble JP, Haines JL, Koller WC, Lyons K, Pahwa R, Stern MB, Colcher A, Hiner BC, Jankovic J, Ondo WG, Allen FH Jr, Goetz CG, Small GW, Mastaglia F, Stajich JM, Mclaurin AC, Middleton LT, Scott BL, Schmechel DE, Pericak-Vance MA, Vance JM (2003) Mitochondrial polymorphisms significantly reduce the risk of Parkinson disease. Am J Hum Genet 72:804–811

    PubMed  Google Scholar 

  • Vance C, Rogelj B, Hortobágyi T, De Vos KJ, Nishimura AL, Sreedharan J, Hu X, Smith B, Ruddy D, Wright P, Ganesalingam J, Williams KL, Tripathi V, Al-Saraj S, Al-Chalabi A, Leigh PN, Blair IP, Nicholson G, De Belleroche J, Gallo JM, Miller CC, Shaw CE (2009) Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science 323:1208–1211

    PubMed  CAS  Google Scholar 

  • Velde CV, Miller TM, Cashman NR, Cleveland DW (2008) Selective association of misfolded ALS-linked mutant SOD1 with the cytoplasmic face of mitochondria. Proc Natl Acad Sci USA 105:4022–4027

    CAS  Google Scholar 

  • Vielhaber S, Kunz D, Winkler K, Wiedemann FR, Kirches E, Feistner H, Heinze H-J, Elger CE, Schubert W, Kunz WS (2000) Mitochondrial DNA abnormalities in skeletal muscle of patients with sporadic amyotrophic lateral sclerosis. Brain 123:1339–1348

    PubMed  Google Scholar 

  • Vijayvergiya C, Beal MF, Buck J, Manfredi G (2005) Mutant superoxide dismutase 1 forms aggregates in the brain mitochondrial matrix of amyotrophic lateral sclerosis mice. J Neurosci 25:2463–2470

    PubMed  CAS  Google Scholar 

  • Vonsattel JP, Difiglia M (1998) Huntington disease. J Neuropathol Exp Neurol 57:369–384

    PubMed  CAS  Google Scholar 

  • Wang J, Xiong S, Xie C, Markesbery WR, Lovell MA (2005) Increased oxidative damage in nuclear and mitochondrial DNA in Alzheimer’s disease. J Neurochem 93:953–962

    PubMed  CAS  Google Scholar 

  • Wang X, Su B, Fujioka H, Zhu X (2008a) Dymanin-like protein 1 reduction underlies mitochondrial morphology and distribution abnormalities in fibroblasts from sporadic Alzheimer’s disease patients. Am J Pathol 173:470–482

    PubMed  CAS  Google Scholar 

  • Wang X, Su B, Siedlak SL, Moreira PI, Fujioka H, Wang Y, Casadesus G, Zhu X (2008b) Amyloid-beta overproduction causes abnormal mitochondrial dynamics via differential modulation of mitochondrial fission/fusion proteins. Proc Natl Acad Sci USA 105:19318–19323

    PubMed  CAS  Google Scholar 

  • 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

    PubMed  CAS  Google Scholar 

  • Wiedemann FR, Manfredi G, Mawrin C, Beal MF, Schon EA (2002) Mitochondrial DNA and respiratory chain function in spinal cords of ALS patients. J Neurochem 80:616–625

    PubMed  CAS  Google Scholar 

  • Wong-Riley M, Antuono P, Ho KC, Egan R, Hevner R, Liebl W, Huang Z, Rachel R, Jones J (1997) Cytochrome oxidase in Alzheimer’s disease: biochemical, histochemical, and immunohistochemical analyses of the visual and other systems. Vision Res 37:3593–3608

    PubMed  CAS  Google Scholar 

  • Wooten GF, Currie LJ, Bennett JP, Harrison MB, Trugman JM, Parker WD (1997) Maternal inheritance in Parkinson’s disease. Ann Neurol 41:265–268

    PubMed  CAS  Google Scholar 

  • Yang Y, Gehrke S, Imai Y, Huang Z, Ouyang Y, Wang J-W, Yang L, Beal MF, Vogel H, Lu B (2006) Mitochondrial pathology and muscle and dopaminergic neuron degeneration caused by inactivation of Drosophila Pink1 is rescued by Parkin. Proc Natl Acad Sci USA 103:10793–10798

    PubMed  CAS  Google Scholar 

  • Yao J, Irwin RW, Zhao L, Nilsen J, Hamilton RT, Brinton RD (2009) Mitochondrial bioenergetic deficit precedes Alzheimer’s pathology in female mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA 106:14670–14675

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Russell H. Swerdlow MD .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2012 Springer Science+Business Media B.V.

About this chapter

Cite this chapter

Lezi, E., Swerdlow, R.H. (2012). Mitochondria in Neurodegeneration. In: Scatena, R., Bottoni, P., Giardina, B. (eds) Advances in Mitochondrial Medicine. Advances in Experimental Medicine and Biology, vol 942. Springer, Dordrecht. https://doi.org/10.1007/978-94-007-2869-1_12

Download citation

Publish with us

Policies and ethics